Category Archives: Embryonic Stem Cells

Confronting IVF: Human Embryos Are Persons With a Right to Life – Walter Bradley Center for Natural and Artificial Intelligence

On Sunday I posted on the Alabama Supreme Courts ruling that frozen embryos are considered children (i.e., minor persons) under Alabama law. The decision is well-reasoned and legally correct, and it highlights the profound ethical issues that accompany in vitro fertilization (IVF) of human beings. In that post, I also commented on Yale neurologist Steven Novellas uninformed opinion on the legal decision.

In that same post, Frozen embryos are not people, Novella offers an opinion on the ethics of IVF that is more thoughtful. I disagree with his conclusion that human embryos are not persons from an ethical standpoint. Ill examine his view step by step, and then provide my own.

Novella:

I [lay] out the core question here when does a clump of cells become a person? Standard rhetoric in the anti-abortion community is to frame the question differently, claiming that from the point of fertilization we have human life. But from a legal, moral, and ethical perspective, that is not the relevant question. My colon is human life, but its not a person. Similarly, a frozen clump of cells is not a child.

What is a potential human?

Unlike many people who deny the personhood of very young human beings, Novella recognizes the obvious scientific fact that human life begins at fertilization and that zygotes/embryos/fetuses/newborns are human beings. They are not potential humans.

A sperm and an egg separately constitute a potential human. But when they unite, the result is a human being from the moment of fertilization. Human beings are not defined by the number of cells in the body be it one cell or 30 trillion cells. A big complex human being is not more human than a small simple human being.

There is no actual debate about this the basic biology of human reproduction was understood in the early 19th century, and any doctor or scientist who denies the humanity of a human being in the womb is either ignorant or deliberately misrepresenting science to advance an ideological agenda. I have explained the scientific fact that human life begins at fertilization here.

And the colon? Novellas invocation of his colon in this debate is nonsense. A colon is an organ its a part of a person, not a whole person. A colon is not a human life. An embryo is a human life, even though he or she is much simpler than Dr. Novellas colon or any other part of an adults body.

This is just basic biology, which Dr. Novella should know.

What is a person?

There are two issues in the IVF debate the question of humanity of the embryo and the question of the personhood of the embryo. They are different questions. The fact that an embryo is a human being is not debatable. But the question Is an embryo a person is another matter, and it is debatable.

What is a person? Novella believes that an embryo is a potential person:

those cells have the potential to become a person. But the potential to become a thing is not the same as being a thing. If allowed to develop those cells have the potential to become a person but they are not a person.

So, according to Novella, what confers personhood on a human being? He states:

The real debate comes down to ethical philosophy and legal theory. How do we balance these various facts:

A human embryo is a human, but not sentient.

Sentience and personhood develops gradually throughout the pregnancy.

Fetuses are dependent on the life of their mother until they develop sufficiently to be viable outside the womb.

Pregnancy is a serious biological process with significant implications for the life of the mother.

Sentience and independence

Novella equates personhood with sentience and independence. Sentience and independence are certainly characteristic of most persons we know (including ourselves), but they are not satisfactory criteria for personhood.

A newborn baby, a person with severe mental handicaps, and a person in a deep coma all lack appreciable sentience and independence but they are undoubtedly persons. Using sentience and independence as fundamental criteria for personhood even implies that temporary unconsciousness such as non-sentience during deep sleep or dependence on others such as being critically ill renders us transient non-persons. In fact, we are persons even when we are non-sentient and dependent on others.

Even more disturbing is the fact that gradations of sentience have long been used to deny basic human rights to categories of people based on real or perceived cognitive differences. An illiterate man is, in a very real sense, less sentient than a literate genius, but is he less of a person?

So what, exactly, is a person? Many different definitions have been offered, but it seems to me that there is one continuous thread that runs through what it means to be a person: a person is a human being who has rights and who is entitled to respect. Certainly, to deny a human being any respect and to deny him all rights is to deny his personhood in a fundamental way.

Most rights of persons are linked to particular acquired skills or milestones in the persons life the right to drive a car, the right to vote, the right to keep and bear arms, etc. Yet there is one right that is not linked to skills or milestones, and it is in fact the right on which all other rights depend the right to life. If I am a person with the right to drive and vote and so on, but not the right to life, my life can therefore be taken from me arbitrarily at the whim of another. Then I really dont have any rights at all, and I am not treated as a person. All other rights are meaningless without the right to life.

The right to life is the indispensable cornerstone of all rights, and the right to life is the cornerstone of personhood. Thus a person is a human being with a right to life.

Are all human beings persons? Or are there two classes of humans?

This is the crux of the debate about IVF, abortion, and other life issues: are all human beings persons with an unalienable right to life? Those of us in the pro-life movement answer that question emphatically in the affirmative. Those who support the destruction of human beings in the womb or in the petri dish, etc., believe there are two classes of human beings: those with the right to life, and those without the right to life. This is the issue at hand.

It is certainly the case that in pregnancy and in the production of frozen embryos there are other rights involved the rights of the mother, the rights of the parents (although, of course, to refer to mother and parents implicitly affirms that the unborn are children, not lumps of cells). In sorting out these various rights the right to bodily autonomy, the right to dispose of property, etc. the right to life always takes precedence.

The right to life is the cornerstone of all rights, and even when the rights of the mother or parents are significant and generally worthy of protection, they do not trump the right to life of the young human being the young person in the womb or in the petri dish or in the nursery.

The implications of IVF

A deeply troubling social and moral issue arises with IVF as well. IVF is the industrial manufacture of human beings. The brave new world that is dawning on us has risks and horrors that should chill us. IVF provides the opportunity to screen embryonic human beings for genetic traits (which is already being done), and this technology can and will be used in the future to breed human beings with certain desired traits obsequious servants, aggressive warriors, attractive sex slaves, and compliant organ donors.

We are learning to mass produce human beings, and although the technology thus far has been used mainly for the sympathetic goal of providing childless couples with children, only a naif would believe that it will end here. The industrial manufacture of human beings opens the door to evil on a scale that is the stuff of nightmares.

IVF is an ethically problematic technology with horrifying implications. If we are to survive this brave new world of mass-produced children, conceived not via conjugal love of a husband and wife in a family but via a pipette in a human assembly line, we must affirm that human life begins at fertilization, and that all human beings are persons with the right to life and deserving of respect. This is the essence of the pro-life movement and the philosophy of human exceptionalism. It is stated most cogently in the Christian view that we are created in Gods image. It is that Image that confers personhood and the unalienable right to life on each of us, from fertilization to natural death.

You may also wish to read: Are IVF human embryos children? A recent court decision Neurologist Steven Novella claims that the Alabama Supreme Court ruling that they are children under the law essentially referenced god The ruling not only did not reference God, it was meticulously based on precedent. So those who seek to remove protection from IVF embryos must lobby for that.

See more here:
Confronting IVF: Human Embryos Are Persons With a Right to Life - Walter Bradley Center for Natural and Artificial Intelligence

In light of the Alabama court ruling, a look at the science of IVF : Short Wave – NPR

Blastocyst illustration. A blastocyst is a hollow ball of cells with a fluid centre formed after several divisions of a fertilised cell (zygote). The inner cell mass (purple) contains the cells that will form the embryo proper, the embryonic stem cells (ESCs). Kateryna Kon/Science Photo Library/Getty Images hide caption

Blastocyst illustration. A blastocyst is a hollow ball of cells with a fluid centre formed after several divisions of a fertilised cell (zygote). The inner cell mass (purple) contains the cells that will form the embryo proper, the embryonic stem cells (ESCs).

Since the first successful in vitro fertilization pregnancy and live birth in 1978, nearly half a million babies have been born using IVF in the United States. Since the first successful in vitro fertilization pregnancy and live birth in 1978, nearly half a million babies have been born using IVF in the United States. Reproductive endocrinologist Amanda Adeleye explains the science behind IVF, the barriers to accessing it and her concerns about fertility treatment in the post-Roe landscape.

For more on IVF success rates, check out the Society for Assisted Reproductive Technology's database.

Questions or ideas for a future episode of Short Wave? Email us at shortwave@npr.org we'd love to hear from you!

Listen to Short Wave on Spotify, Apple Podcasts and Google Podcasts.

Listen to every episode of Short Wave sponsor-free and support our work at NPR by signing up for Short Wave+ at plus.npr.org/shortwave.

This episode was produced by Berly McCoy and Rebecca Ramirez. It was edited by Brit Hanson and Rebecca Ramirez. Brit checked the facts. The audio engineer was Josh Newell.

See the original post:
In light of the Alabama court ruling, a look at the science of IVF : Short Wave - NPR

Runx1+ vascular smooth muscle cells are essential for hematopoietic stem and progenitor cell development in vivo – Nature.com

A subpopulation of subaortic mesenchyme in the AGM co-expresses NG2 and Runx1

We examined the expression of PC/vSMC markers in the dorsal aorta of E10.5 and E11 mouse embryos. Wholemount immunostaining and immunohistochemistry on frozen sections were performed using PC/vSMC markers NG2 or SMA with CD31, an endothelial and HSPC marker (TableS1). Imaging analysis showed that NG2+SMA+CD31- vSMCs surround NG2-SMA-CD31+ endothelial cells (Figs.1a, S1a, b), confirming previous reports27. Further to its expression in hematopoietic and hemogenic endothelial cells, Runx1 was also detected in the sub-aortic mesenchyme22,23. Therefore, we hypothesised that at least some of these cells also express NG2. We first confirmed that both intra-aortic hematopoietic cell clusters (IAHCs) (Fig.1b, stars) and hemogenic endothelial cells (Fig.1b, arrowheads) are Runx1+; we also identified a subpopulation of NG2+ PC/vSMCs, mainly located in the ventral aspect of the dorsal aorta, that also express Runx1 (Fig.1b, arrows). Other Runx1+ cells in the perivascular area do not express NG2 (Fig.1b). Finally, we confirmed our recent study28 that some cells around the notochord express NG2 in the trunk (Fig.S1a, circle). However, these peri-notochord cells do not express SMA, CD31 (Fig.S1a, circle) nor Runx1 (Fig.S1ef). To confirm the presence of NG2+Runx1+ cells in the E11 AGM, we used Runx1-IRES-GFP mouse embryos29. In these GFP knock-in mice, GFP intensity correlates with Runx1 expression level. Flow cytometric analysis showed the presence of a distinct population of NG2+Runx1(GFP)+ cells in the AGM (Fig.1c). These cells first appear at E10, in line with the presence of Runx1 in mesenchymal cells30 and importantly, their frequency peaks at E10.5 (Fig.1d). Together, these data show that in the AGM, a subset of the sub-aortic mesenchyme expresses both NG2 and Runx1 and that the highest frequency of these cells coincides with the onset of HSC generation at E10.5.

a Three-dimensional (3D) wholemount immunostaining with SMA, CD31 and NG2 of E10.5 (3138 somite pairs (sp)) WT dorsal aorta; b NG2 and Runx1 expression on single plane wholemount WT E10.5 sections. NG2+Runx1+vSMCs (arrows), hemogenic endothelial cells (arrowheads) and intra-aortic hematopoietic clusters (IAHCs, stars) (TableS1); c Representative example of flow cytometric analysis of NG2+Runx1(GFP)+ (green box) in E10.5 Runx1-IRES-GFP AGM and E10.5 WT control. d Percentages of NG2+Runx1(GFP)+ cells in E9 (21-25sp) body (n=6), E10/E10.5/E11 AGMs (n=8/7/7), N=5, Kruskal-Wallis and Dunns post-hoc test. e Representative examples of wholemount 3D-images showing SMA, CD31 and NG2 in E10.5 cKO dorsal aortae; f SMA, Runx1 and CD31 immunofluorescence of E11 WT and cKO transversal frozen sections; n=WT/cKO: 2/2, N=2. g cKit and CD31 wholemount 3D-images in E10.5 WT and cKO AGM; h Number of intra-aortic hematopoietic clusters (IAHCs) in E10.5 AGM; n=WT/KO: 5/4, N=4. Number of colony forming unit-culture (CFU-C) in i E10.5 (31-38sp) AGM; n=WT/HET/KO: 14/10/5 embryos; N=7 and j E11 (4352sp) AGM; n=WT/HET/KO: 22/8/19 embryos; N=11; one-way ANOVA and Tukeys post-hoc test (TableS2). k Percentages of donor cell chimerism 4-months post-transplantation of 6 E11 WT (NG2+/+;Runx1fl/+or NG2+/+;Runx1fl/fl), 7 HET (NG2-Cre;Runx1fl/+) and 6 cKO AGMs (NG2-Cre;Runx1fl/fl) into sub-lethally adult irradiated recipients (1xAGM cells transplanted/recipient; N=4). Each dot represents one recipient. Mice are reconstituted when 5% donor cells are found in the host peripheral blood (dashed line); one-tailed Z score test for two population proportions (TablesS3 and S4). For wholemount staining in a, b, e, g: WT/cKO (N=6/4): SMA (n=9/7), CD31 (n=10/7), cKit (n=3/2), NG2 (n=3/1) and WT Runx1 (n=4) in 3 distinct combinations (TableS1). D = dorsal, V = ventral. N = number of independent experiments; n = number of biological samples (embryos). All data are presented as mean valuesSEM. Source data for d, h, i, j and k are provided as a Source Data file.

Runx1 deletion in endothelial cells impairs HSC emergence in the AGM24,25,26. However, the effect of Runx1 deletion in PC/vSMCs on hematopoiesis in vivo is still unknown. To address this, we examined conditional knock-out (cKO) NG2-Cre;Runx1fl/fl mouse embryos. In previous studies, the NG2-Cre mouse strain revealed a role for pericytes in supporting both fetal liver and adult bone marrow HSC maintenance31,32. Our data shows that E10.5 and E11 cKO embryos do not exhibit visible vascular abnormalities. This was confirmed by the normal expression of CD31, SMA and NG2 (Figs.1e, f, S1c, d) in the AGM. In contrast, SMA+Runx1+ PC/vSMCs with low expression of Runx1 were reduced in the cKO dorsal aorta compared to WT littermate controls (Figs.1f, S1eg). CD31+Runx1+ endothelial cell number and frequency was also decreased (Fig.S1g). Furthermore, CD31+cKit+ IAHC numbers were significantly reduced by three-fold (p=0.02) (Fig.1g, h). Hematopoietic progenitor (HP) assays were performed to test if hematopoietic function was affected. All HP numbers were significantly reduced in cKO AGMs at both E10.5 (Fig.1i, TableS2) and E11 (Fig.1j, TableS2). To test whether definitive HSCs were also affected, we performed HSC assays in vivo. At 1- and 4-months post-transplantation of AGM cells into sub-lethally irradiated mice, chimerism and multilineage reconstitution were examined by flow cytometry in the peripheral blood. Compared to the WT littermate control group, in which 66.7% (4 out of 6) recipients were reconstituted, only 14.3% (1 in 7, p=0.025) and 16.7% (1 in 6, p=0.040) mice injected with heterozygous or homozygous cKO AGMs, respectively, were reconstituted over the long term (Fig.1k, TablesS34). These findings indicate that the absence of Runx1 in aortic NG2+ cells impairs HSC generation and/or maintenance and HP development in the AGM.

To test whether NG2+ cells contribute directly to hematopoietic lineages, we isolated NG2+ and NG2+Runx1(GFP)+cells from E11 WT and Runx1-IRES-GFP AGMs, respectively, and seeded them in methylcellulose. In parallel, NG2- or NG2-cKit+ cells were sorted as controls. HPs were exclusively found in the NG2- cell fractions. Neither NG2+ cells (Fig.S2a) nor NG2+Runx1(GFP)+cells (Fig.S2b) gave rise to hematopoietic cell colonies in vitro (TableS5). To further assess whether NG2+ cells are hematopoietic precursors, we crossed NG2-Cre mice with a knock-in reporter mouse line in which tdTomato is preceded by a transcriptional stop flanked by two loxP sites under the Rosa26 promoter. In these mice, NG2+ cells and their progeny are tdTomato+. E11 AGM-derived tdTomato+ and tdTomato- cells were sorted and seeded in methylcellulose. HPs were only found in the tdTomato- cell fraction (Fig.S2c, TableS5) reinforcing the observation that NG2+ cells and their progeny do not contribute to hematopoietic lineages at this stage. Flow cytometric analysis confirmed the presence of tdTomato in a subset of NG2+ cells in the E11 AGM (Fig.S2d), validating our mouse model, while no overlap was found between tdTomato and CD45, a hematopoietic cell marker (Fig.S2e). We next performed immunohistochemistry on NG2-Cre;tdTomatofl/+ frozen sections and confirmed the expression of tdTomato in a subset of SMA+ cells (Fig.S2f) in the E11 AGM. CD31+ cells did not express tdTomato (Fig.S2g). Further analysis revealed that cells expressing hematopoietic markers F4/80 and CD45 do not co-express NG2 nor SMA (Fig.S2h). Together, these data indicate that NG2+ cells do not contribute to the AGM HSPC pool and suggest that NG2+Runx1+ PC/vSMCs act as a supportive niche to maintain hematopoietic activity in the AGM.

In the early developing embryo, HSPCs reside in other intra-embryonic and extra-embryonic hematopoietic organs such as the head, fetal liver (FL), placenta and yolk-sac (YS). Flow cytometric analysis of these organs harvested from Runx1-IRES-GFP mouse embryos also confirmed the presence of NG2+Runx1(GFP)+ cells (Fig.S3a, b). We next performed in vitro HP functional assays with cells harvested from all organs and genotypes of NG2-Cre;Runx1fl at both E10 and E11 developmental stages. No significant differences were found when comparing the total CFU-C numbers between genotypes in most organs (Fig.S3c, d, TablesS67). A significant increase of total number of CFU-C was observed in E10 AGM in both heterozygous and cKO mouse embryos (Fig.S3c). When analyzed individually, a significant increase in the number of erythroid colonies was detected in the cKO compared to WT littermate (p=0.0149) (TableS6). Likewise, a 2.8-fold increase in the number of erythroid colonies was detected in the E11 cKO head compared to the WT littermate (p=0.01), while the total number of CFU-C in the E11 head remained unchanged (TableS7). Moreover, we found a significant decrease in both CFU-GM (p=0.016) and CFU-GEMM (p=0.039), between WT and cKO YS (TableS7), possibly due to the defect found in the E11 AGM.

To test whether HSC activity increases in the FL due to the possible migration of AGM HSCs, E11 FL cells from all genotypes were transplanted into sub-lethally irradiated recipient mice. Neither the donor chimerism nor the percentage of reconstituted mice by donor cells showed changes between the groups (Fig.S3e). Compared to NG2+/+;Runx1fl/+ WT littermates, in which 70% of recipients (7 out of 10) were reconstituted, mice injected with NG2-Cre:Runx1fl/+ heterozygous or NG2-Cre:Runx1fl/fl cKO E11 FL showed similar reconstitution over the long term, with 67% (2 out of 3, p=0.348) and 60% (3 out of 5, p=0.421) reconstituted mice, respectively (Fig.S3e, TablesS34). Since the deletion of Runx1 in NG2+ cells only affects HSPCs in the AGM, immunohistochemistry on WT embryonic head and placenta was performed to localise NG2+Runx1+ cells. The rare NG2+Runx1+ double positive cells identified did not seem to be perivascular (Fig.S3f, stars). In line with this observation, we found that Runx1 and SMA do not overlap when NG2 and SMA were expressed in PC/vSMCs (Fig.S3f, arrowheads). Instead, the head contains few NG2+SMA- that are F4/80+, suggesting that NG2+Runx1+ cells are macrophages (Fig.S3f, arrowhead). Overall, our data shows that the deletion of Runx1 in NG2+ cells only affects selective HSPC subsets in non-AGM hematopoietic organs in the E11 mouse embryo.

To better understand the role of Runx1 in the HSC-generating microenvironment, single-cell RNA-sequencing (scRNA-seq) on NG2+/+;Runx1fl/+ E11 AGM was performed. We used graph-based clustering and known marker distribution to define and investigate the gene expression profiles of various populations that reside in the E11 AGM and identified eight populations of interest (Fig.2a, b). The co-expression of Cspg4 (NG2) and Acta2 (SMA) in the PC/vSMC population was confirmed (Fig.2c). This population is also enriched in Rgs5, Pdgfrb and Pdgfra in line with our previous work28, and a subset of these cells express Runx1 (Fig.2c, d), confirming our imaging and flow cytometric analysis. The expression of Mcam (CD146 or S-ENDO1), a pericyte/vSMC precursor marker recently identified in a subset of NG2+ cells in the E11 AGM21 and upregulated in AGM hematopoiesis supportive stromal cell lines19, was detected in a subset of PC/vSMCs, partially overlapping with Runx1+ cells (Fig.2c, d). However, Mcam was mainly enriched in endothelial cells (ECs) and also in subpopulations of hemogenic endothelial cells, including those entering endothelial-to-hematopoietic transition (HEC/EHT), IAHCs and SNS cells (Fig.2d), confirming published work including ours28,33. Immunostainings with CD146 and CD31 on E11 WT AGM frozen sections further validated our sequencing analysis at the protein level: both CD31+ endothelial cells (Fig.2e, f, arrows) and SMA+ PC/vSMCs (Fig.2f, stars) are CD146+. Importantly, Pecam-1 (CD31) expression in PC/vSMCs was low/negative in our scRNA-seq data (Fig.2c, d), in line with our immunohistochemistry, confocal imaging, and our recent published work28. Other genes expressed by hematopoietic and hemogenic/endothelial cells such as Adgre1 (F4/80), Mrc1 (CD206), Cdh5 (VE CADHERIN), Tek (TIE-2), CD34, CD93, Pdgfb, Sox7, Sox17, Sox18, Gfi1b, and Itga2b (CD41) were not expressed in PC/vSMCs (Fig.2d). These genes were used to distinguish populations of macrophages (MPs), IAHCs, HEC/EHT, and ECs (Fig.2ad). Erythroid cellsand erythroid progenitors (Ery/EryP; Gypa/CD235+), SNS (Gata3+) and skeletal muscle progenitors (SkMP; MyoD1+, Cdh15+) were also identified (Fig.2ad). Kit was expressed in all IAHCs and in a subset of PC/vSMCs, while Kit expression in HEC/EHTs was low (Fig.2d). Altogether, these data show that we successfully captured multiple cell types that comprise the E11 AGM, including a population of Runx1+ PC/vSMCs which constitutes 19.7% of all NG2+Acta2+ PC/vSMCs cells. Furthermore, the transcriptome of Cspg4+Runx1+ non-hematopoietic non-endothelial PC/vSMCs was found to partially overlap with that of the Cspg4+Mcam+ PC/vSMC precursors previously described21.

a t-SNE plot highlighting eight populations of interest identified in the E11 WT AGM. Each dot represents one cell and colours represent cell clusters as indicated. The number of cells in each population is shown in brackets. MP (macrophages); Ery/EryP (erythroid/progenitors); IAHC (intra-aortic hematopoietic clusters); HEC/EHT (hemogenic endothelial cells including those that enter endothelial-to-hematopoietic transition); EC (endothelial cells); SNS (sympathetic nervous system); SkMP (skeletal muscle progenitors), PC/vSMC (pericytes/vascular smooth muscle cells, NG2+Acta2+). Other cells (OC) are coloured in grey. b t-SNE plot highlighting the eight populations identified after excluding all other (grey) cells. c Zoom into PC/vSMC cluster (black rectangle) further show the presence or the absence of selected genes that characterise this population and confirms the presence of Runx1 in a subset of cells. d Violin plots showing distribution of expression for selected genes that contributed to the identification of cell clusters. Immunohistochemistry on frozen E11 WT sections stained with eCD146/CD31/DAPI and fCD146/SMA/DAPI, n=2 samples tested, N=2 independent experiments. Arrows: vascular cells, asterisks: perivascular cells. DA: dorsal aorta, CV: cardinal veins, NC: notochord. Source data for e (first column,20X) is provided as a Source Data file.

Our scRNA-seq analysis revealed that not all Cspg4+Runx1+ cells in the E11 AGM express Acta2 (Fig.3a, b). We therefore investigated if Cspg4+Runx1+Acta2 cells are PC/vSMCs which had not yet acquired Acta2 expression. Differential expression analysis of Acta2+ versus Acta2 cells within the NG2+Runx1+ cell population in the WT AGM revealed that markers of sclerotome-derived vSMCs such as Sox9, Pax1, Pax9 and Col2a134 are among the highest upregulated genes in Cspg4+Runx1+Acta2 cells (Fig.3c). In contrast, Cspg4+Runx1+Acta2+ cells are enriched in genes that identify more mature pericytes such as Acta2, CD248, Mcam, Rgs5 or Pdgfrb (Fig.3c), some of which are potential Runx1 target genes (star). Pdgfra and Ptn genes were recently associated with Runx1+ subaortic (non-smooth muscle) mesenchymal cells with possible role in hematopoiesis in the E10.5 AGM of the mouse embryo35. Our scRNA-seq analysis show that, in E11 AGM, Pdgfra and Ptn are also expressed in Cspg4+Runx1+ cells with no significant difference between Acta2+ and Acta2 (Fig.3c). Further analysis showed that Gene Ontology (GO) biological processes significantly enriched in Cspg4+Runx1+Acta2+ cells include smooth muscle cell chemotaxis and migration, collagen-activated signalling pathway, neural crest cell differentiation and regulation of BMP signalling (Fig.3d), previously shown by our laboratory to control in vivo HSPC generation in the mouse AGM28. In Cspg4+Runx1+Acta2 cells, significantly enriched GO biological processes include mesenchymal stem cell differentiation and cartilage and bone development (Fig.3e), consistent with the sclerotome origin of these cells.

a t-SNE plots showing the distribution of Runx1 and Acta2 expression in NG2+Runx1+ cells in the WT E11 AGM after excluding all other (grey) cells found in the Fig.2a. b Zoom into NG2+Runx1+ cluster (black rectangle) shows the presence or the absence of Acta2. c Heatmap showing the expression of Cspg4 and Runx1 and 15 selected genes out of 25 top significantly upregulated genes in WT NG2+Runx1+Acta2+ cells (upper half) and NG2+Runx1+Acta2- cells (bottom half) at single cell level; *Runx1 potential target genes. Pdgfra and Ptn genes were next added to inform their expression in both populations. Barplot of fold enrichment for selected GO biological processes significantly overrepresented in genes significantly upregulated in both dWT NG2+Runx1+Acta2+ and eNG2+Runx1+Acta2- cells. f t-SNE of WT E11 AGM cells, overlaid with principal pseudotime curve inferred by Slingshot, predicting a lineage from NG2+Runx1+Acta2- cells to NG2+Runx1+Acta2+ cells. g WT NG2+Runx1+cells arranged in pseudotime (x-axis) based on the inferred curve. Y-axis represents log normalised gene expression.

Indeed, PC/vSMCs in the AGM have been shown to originate from the sclerotome and display markers of this compartment at least during the early phases of mural cell recruitment36. A recent study showed that the maturation of sclerotome-derived vSMCs in the mouse AGM depends on a transcriptional switch from a sclerotome signature with the repression of Pax1, Scx and Sox9, and activation of Acta2 and other vSMC genes34.

To test whether NG2+Runx1+Acta2 cells follow a maturation trajectory towards Cspg4+Runx1+Acta2+ vSMCs, we performed cell lineage inference with Slingshot, a trajectory inference method for scRNA-seq data that can incorporate knowledge of developmental markers. Having defined a cluster of Cspg4+Runx1+Acta2 cells as an origin, Slingshot infers a cell lineage and constructs a pseudotime curve representing that lineage (Fig.3f, arrow). Gene expression along pseudotime shows that sclerotome markers such as Sox9, Pax1 and Pax9 are gradually downregulated while markers of mural cells such as Acta2, Rgs5, Pdgfr, Cnn1, Mcam and CD248, are gradually upregulated in an inferred transition from Cspg4+Runx1+Acta2 to Cspg4+Runx1+Acta2+ cells (Fig.3g). Our scRNA-seq analysis shows that Cspg4+Runx1+ AGM cells display a sclerotome-derived vSMC transcriptomic profile.

We next explored the impact of Runx1 deletion in the hematopoietic niche and its possible effect on PC/vSMCs by performing scRNA-seq of NG2-Cre;Runx1fl/fl cKO E11 AGMs (Fig.4a, b). Cell populations were defined in a similar way to the WT AGM by using graph-based clustering and known marker distribution. This comparison revealed changes in the proportions of the different cell types between WT and cKO AGM, including a significant reduction in the proportion of cells associated with clusters 2 (Ery/EryP), 3 (IAHC), 6 (SNS), and 7 (SkMP) (Fig.4c).

a t-SNE plot showing eight populations of interest found in the E11 cKO AGM. Each dot represents one cell and colours represent cell clusters as indicated. MP(macrophages); Ery/EryP(erythroid/progenitors); IAHC (intra-aortic hematopoietic clusters); HEC/EHT (hemogenic endothelial cells including those that enterendothelial-to-hematopoietic transition),EC (endothelial cells); SNS (sympathetic nervous system); SkMP(skeletal muscle progenitors); PC/vSMC (pericytes/vascular smooth muscle cells, NG2+Acta2+). Other cells (OC)are coloured in grey. The number of cells in each cluster is shown in brackets. b t-SNE plot highlighting the eight populations identified after excluding all other (grey) cells. c Percentage of single live cells found in each E11 AGM sample (cell number/total cells) defined by scRNA-seq in WT (full bars) and cKO (empty bars) AGMs. Colours and numbers correspond to each population defined in a; chi-squared two-tailed test was used for comparison. d Barplot of fold enrichment for selected GO biological processes significantly overrepresented in genes significantly downregulated in cKO PC/vSMCs compared to their WT counterparts. Heatmap of ligand-receptor interactions inferred by NicheNet from e WT and f cKO E11 AGM cells. Colour represents the interaction potential score between the 10 top-ranked ligands expressed in ECs and their inferred targets expressed in PC/vSMCs. Ligands and receptors are ordered by hierarchical clustering. g Scatter plots of AUC vs log10(FDR) showing downregulated genes associated with selected GO terms in cKO PC/vSMCs. Red dots represent significantly downregulated genes (FDR<0.05); dashed line shows FDR = 0.05. Gene labels with red borders represent potential Runx1 target genes.

Changes in gene expression between WT and cKO Cspg4+Runx1+ cells were first investigated. We found that genes significantly downregulated in cKO Cspg4+Runx1+ cells were mainly associated with biological processes including translation, oxidative phosphorylation, cellular response to stress and mitochondria-related function (Fig.S4ac). As deletion of Runx1 may have also affected Runx1 PC/vSMCs, the transcriptome of all cKO Cspg4+Acta2+ PC/vSMCs with their WT counterpart was compared. Genes significantly downregulated in cKO Cspg4+Acta2+ PC/vSMCs had significant enrichment of biological processes including translation, smooth muscle cell differentiation, cytoskeleton, vasculogenesis and cell communication (Fig.4d). One pathway essential to vasculogenesis is PDGF-B/PDGFR; we therefore applied NicheNet on our WT scRNA-seq data to predict ligand-receptor interaction between ECs and PC/vSMCs, focusing on PDGF-B-related genes. The highest scoring predicted interaction was between Pdgfb, a growth factor released by ECs, and Nrp1 (Fig.4e), a receptor known to control the differentiation/recruitment of mesenchymal stem cells and the stimulation of smooth muscle cell migration37,38.

The interaction between Pdgfb and Pdgfrb was also amongst the highest scoring interactions in both WT (Fig.4e) and cKO (Fig.4f). Additional interactions involve Edn, Tgfb or Bmp pathways, previously associated with a role in AGM hematopoiesis39,40. Interestingly, in cKO ECs, Pdgfa, another gene from the PDGF family, was no longer in the top 10 ranking ligands (Fig.4f) possibly due to the downregulation of Pdgfra in cKO PC/vSMCs (Fig.4f). Other genes including Des, Angpt1, Gsk3b, Tcf21, Col1a1, Pcna, Ccnd3 and Mcm7, potential Runx1 downstream target genes41, were also significantly downregulated (Fig.4g, red boxes). The reduction inCol1a1 expression suggests changes in the gene profile of the extracellular matrix (ECM). Indeed, additional ECM related genes were significantly downregulated in the cKO PC/vSMCs, such as Sparcl1, Col3a1 and Col5a1 (Fig.4g). Collectively, these data show that the genetic programme of PC/vSMCs in cKO AGM is modified upon Runx1 deletion and this involves changes in molecules that constitute the ECM of the aortic wall.

Endothelial cells share the same basement membrane with PC/vSMCs42. This, coupled with the transcriptomic changes in the cKO PC/vSMCs described above, suggest that the genetic programme of the adjacent ECs may have also been altered by Runx1 deficiency in NG2+ cells. Although the number of endothelial cells in the NG2-Cre:Runx1fl/fl cKO did not significantly change (Fig.4a) and the formation of the dorsal aorta appeared to be unaffected (Fig.1e), we investigated transcriptomic changes in ECs that could affect their function in vivo. As before, we performed differential expression analysis, followed by overrepresentation analysis on genes significantly downregulated in cKO ECs (Fig.5a). Multiple GO biological processes were significantly overrepresented in these genes, with many related to EC development and angiogenesis; proliferation, migration and differentiation; response to hypoxia and fluid shear stress; as well as smooth muscle cell or mesenchymal cell development and hematopoiesis (Fig.5a). Interestingly, we found that Sox18 was the most downregulated gene in cKO ECs (Fig.5b). Col4a1, the most abundant extracellular matrix associated gene, known to co-localise with Sox18 in ECs in the mouse embryo43, was also found within the top 25 downregulated genes (Fig.5b). Sox18 and Col4a1 were the most downregulated genes associated with the blood vessel development GO term, while other gene expression including Cdh5, Pecam1, Sox17, Pdgfb, MCam and Notch were also affected.

a Barplot of fold enrichment for selected GO biological processes significantly overrepresented in genes significantly downregulated in cKO ECs compared to their WT counterparts. b Scatter plots of AUC vs log10(FDR) showing downregulated genes associated with selected GO terms including blood vessel development and mesenchymal cells and vSMCs in cKO ECs. Red dots represent significantly downregulated genes (FDR<0.05); dashed line shows FDR=0.05. Sox18 and Ctnnb1 expression in WT ECs in both scRNA-seq (c, EC zoom and t-SNE plots) and bulk RNA-seq post-sort (d, TPM). e Scatter plots of AUC vs log10(FDR) showing downregulated genes associated with selected GO terms including the basement membrane and extracellular matrix in cKO ECs. Red dots represent significantly downregulated genes (FDR<0.05); dashed line shows FDR=0.05. Selected genes that were altered in cKO ECs ine are shown in WT ECs in both scRNA-seq (f, ECand HEC/EHT zoom and t-SNE plots) and bulk RNA-seq post-sort (g, TPM). TPM: transcript per Million mapped reads values.

Genes associated with cell adhesion, regulation of smooth muscle cell proliferation and differentiation, along with mesenchyme development such as Sox18 and Ctnnb1 were also significantly downregulated in cKO EC (Fig.5b, arrow). We confirmed that both Sox18 and Ctnnb1 are expressed by ECs in our single cell datasets (Fig.5c) and next validated their expression in NG2-PDGFR-ckit-CD45-CD31+ Runx1- purified ECs from E11 Runx1-IRES-GFP AGMs (Figs.5d, S5a).

Some significantly downregulated genes associated with blood vessel development such as Loxl2, Hspg2, Col4a2, Col15a1 and Col18a1 (Fig. 5b)are also known to be associated to the ECM. Further analysis of endothelial extracellular matrix encoding genes previously described44 revealed that most of these genes were also significantly downregulated in cKO ECs (Fig.5e). The expression of these genes in WT ECs at single-cell level (Fig.5f) was confirmed post-sort at population-level (Fig.5g) with most genes being highly expressed in ECs only. One of them was Sparc (Fig.5e blue arrow, Fig.5g), a central ECM secreted Ca2+-binding glycoprotein that interacts with many other ECM proteins including Col1 and Col445,46. Among the SPARC family, Sparcl1 (Sparc-like 1), known to bind to Col147, was also found to be significantly downregulated in cKO ECs (Fig.5b, c). Together, these analyses show that Runx1 deficiency in NG2+cells leads to significant transcriptomic changes in endothelial cells including extracellular matrix related genes. We did not detect transcriptional changes in the NG2-Cre;Runx1fl/fl cKO HEC/EHT cell cluster, although this observation is inconclusive due to the low number of cells captured.

Transcriptomic changes in vascular and perivascular cells may have also affected IAHCs. As hematopoietic cells are highly heterogeneous and progenitors were significantly affected (Fig.1), we first explored WT IAHCs in more detail. Previous studies showed that IAHCs are composed of both Runx1+ and Runx1- cells28,48,49 and we were able to confirm this by flow cytometry in Runx1-IRES-GFP AGMs (Fig.S5a). We also confirmed the expression of Runx1 in HEC/EHT and its absence in ECs by flow cytometry in Runx1-IRES-GFP E11 AGMs (Fig.S5a), in line with published work50. To validate their cell identity, we next purified and sequenced 243 Runx1 (GFP)+ and 27 Runx1 (GFP)- IAHCs (NG2-PDGFR- CD31+ckit+) as well as 5822 EC (Runx1-) and 248 HEC/EHT (Runx1+) cells from NG2-PDGFR- ckit- CD45- CD31+ E11 Runx1-IRES-GFP AGMs (Fig.S5a) and performed bulk RNA sequencing (RNA-seq). The purity of the sort was first confirmed (Fig.S5b). While CD45 antibody was not used to isolate IAHCs (Fig.S5a), our bulk RNA-seq data (Fig.S5b) show that not all IAHC cells express Ptprc (CD45) in line with previous studies48,49,51, and seems to be present only when Runx1 is expressed. Next, the identity of all sorted cell populations based on the expression of genes known to be expressed in these cells15,35,48 was confirmed (Fig.S5c). Interestingly, the transcriptomic profile of Runx1 (GFP)+ and Runx1(GFP)- sorted IAHCs appears to be distinct. While CD34, Gata2, Lmo2, Etv6, and Eglf7 are expressed in both Runx1(GFP)+ and Runx1(GFP)- IAHCs at various levels, Adgrg1, Gfi1, Myb and CD44 are mainly found in Runx1(GFP)+ IAHCs (Fig.S5c). Instead, as they also express Tek, Kdr, Eng, Esam and Gata2 (Fig.S5c, d), Runx1(GFP)- IAHCs are at the transcription level, closer to type-1 pre-HSCs52,53 or to recently described CD31+ckithighGata2medium IAHCs that are Runx1-Ptprc-48 with possible (micro)-niche role54. Our analyses confirm the heterogeneity of Runx1(GFP)+/-CD31+C-KIT+IAHCs in the E11 AGM at both protein and transcriptomic levels, and indicate that most IAHC cells captured in our full/unsorted AGM scRNA-seq are Runx1-.

To explore transcriptomic changes between WT and cKO IAHCs, differential expression analysis followed by overrepresentation analysis on genes significantly downregulated in cKO IAHCs was carried out (Fig.6a). Several GO biological processes were significantly overrepresented in these downregulated genes, including ribosome assembly processes, regulation of translation, RNA transport and localisation, and others such as response to DNA damage, gene expression and cellular processes (Fig.6a). In line with this, we found that the top 25 significantly downregulated genes in cKO IAHCs were mostly ribosomal protein coding genes from both Rps and Rpl families. Other genes in the top 25 are known to be required for transcriptional or translational initiation such as Btf3, Pabpc1 and Bclaf1 (Fig.6b). Interestingly, one of the top significantly downregulated genes in the cKO was Sox18 (Fig.6b, arrow), previously reported to be expressed in both IAHCs and ECs in the mouse AGM55 and confirmed here by our WT scRNA-seq data (Fig.2d). Furthermore, Sox18 has been transiently detected during early hematopoiesis in a model of embryonic stem cell differentiation in vitro, controlling early HP proliferation and maturation56. In line with this, further GO analysis revealed that Sox18 is associated with cellular processes including cell maturation, cell differentiation and regulation of stem cell proliferation (Fig.6b). The latter two GO terms are also associated with other genes significantly downregulated in cKO IAHCs such as Hmgb2, encoding a chromatin-associated non-histone protein involved in transcription and chromatin remodelling (Fig.6b). This transcriptomic analysis shows that the deletion of Runx1 in NG2+ PC/vSMCs within the AGM niche significantly alters the genetic programme of IAHCs.

a Barplot of fold enrichment for selected GO biological processes significantly overrepresented in genes significantly downregulated in cKO HSPCs compared to WT HSPCs. b Scatter plot of AUC (representing strength of downregulation) vs log10(FDR), showing the top 25 significantly downregulated genes (red circles) in cKO HSPCs. Scatter plots of AUC vs log10(FDR) highlighting downregulated genes associated with Gene Ontology (GO) biological processes. Red dots found above the dashed line (corresponding to FDR=0.05) represent significantly downregulated genes (FDR<0.05).

Despite the decrease in HPs and HSCs in cKO AGM, NG2-Cre;Runx1fl/fl mice are born with no obvious defects and develop into adulthood. Because of this, we sought to explore the effect of Runx1 deletion in NG2+ PC/vSMC on adult HSPCs. The presence of these progenitors in the adult bone marrow (BM) of mutant mice was analyzed by flow cytometry and compared to WT mice. No significant differences were found in either Lin-Sca1+cKit+ (LSK) (Fig.7a, b) nor LSK CD150+CD48-(SLAM) cell frequencies (Fig.7c, d) between cKO mice and WT controls. We performed HP assays and found that the frequencies of hematopoietic cell colonies were similar in all mutants and WT littermates (Fig.7e, TableS8). To assess the capacity of these cells to reconstitute hematopoiesis in vivo, 5105 bone marrow cells harvested from all genotypes were transplanted into sub-lethally irradiated WT mice recipients. Compared to the control group in which 62.1% (18 out of 29) mice were reconstituted, mice injected with NG2-Cre;Runx1fl/+ or NG2-Cre;Runx1fl/fl BM cells showed a significant reduction in the long-term reconstitution potential, with only 27.3% (3 out of 11, p=0.024) and 20% (4 out of 20, p=0.002) of transplanted mice being reconstituted respectively (Fig.7f, TableS3). In addition, the percentage of donor chimerism was significantly reduced in the cKO group. On average, the donor chimerism with WT cells was 33% compared to the 16% and 9% observed when BM cells from NG2-Cre;Runx1fl/+heterozygous and NG2-Cre;Runx1fl/fl cKO (p=0.002) were injected respectively (Fig.7f, TableS4). The remaining HSCs in the mutant NG2-Cre;Runx1fl/+ and NG2-Cre;Runx1fl/fl adult BM are multilineage, showing similar contributions of donor cells to myeloid or lymphoid cell compartments (Fig.7g), and self-renew (Fig.7h). Interestingly, no NG2+Runx1(GFP)+ cells were detected in adult Runx1-IRES-GFP BM hematopoietic niches (Fig.7i), suggesting that they are exclusive to the embryo and that the BM hematopoietic defect found in adults is developmentally driven.

a, bRepresentative plots and percentages of Lin-Sca1+cKit+ (LSK) and c, dLSK CD150+CD48-(SLAM) bone marrow (BM) cells by flow cytometry of WT/ NG2+/+;Runx1fl/+,NG2+/+;Runx1fl/fl (n=9), HET NG2-Cre;Runx1fl/+ (n=4) and cKO NG2-Cre;Runx1fl/fl (n=4) adult mice is shown. e Colony-forming unit-culture (CFU-C) numbers per 104 adult BM cells; n=WT/HET/cKO: 13/7/8 mice. N=7 independent experiments. Data are meanSEM (TableS8). f Hematopoietic stem cell repopulating potential and donor chimerism of WT and mutant BM cells in vivo. 5105 BM donor WT, HET and cKO cells were injected into 29, 11 and 20 Ly5.1 HET recipients, respectively, with 18, 3 and 4 found to be reconstituted respectively (Table S3, p=0.024 (WT/HET) and p=0.002 (WT/cKO) by Z score test for 2 population proportions). Mice are reconstituted when 5% donor cells are found in the host peripheral blood; p=0.002 (WT/cKO) by Kruskal-Wallis and Dunns post-hoc test (TableS4). g Histograms showing the contribution of CD45.2+CD45.1- donor cells to myeloid cells (CD11b+Gr1+/-), B cells (CD19+) and T cells (CD4/8+) in all reconstituted host mice from (f). (n=WT/HET/cKO=18/3/4), p=0.019 (WT/HET) for B cells by one-way ANOVA and Tukeys post-hoc test. h BM cells from selected reconstituted primary recipients (found in f) were transplanted into multiple irradiated secondary recipients. Mice are reconstituted when 5% donor cells are found in the host peripheral blood (TableS34). i Representative flow cytometric analysis plot of NG2 in Runx1-IRES-GFP adult BM (n=6). All data are presented as Mean values+/-SEM. N=number of independent experiments; n = number of biological samples. Source data for b, d, e, f, g and h are provided as a Source Data file.

Follow this link:
Runx1+ vascular smooth muscle cells are essential for hematopoietic stem and progenitor cell development in vivo - Nature.com

Alabama’s biggest hospital to suspend transfer of embryos after court ruling – ABC News

This page either does not exist or is currently unavailable.

From here you can either hit the "back" button on your browser to return to the previous page, or visit the ABCNews.com Home Page. You can also search for something on our site below.

STATUS CODE: 500

The rest is here:
Alabama's biggest hospital to suspend transfer of embryos after court ruling - ABC News

U.S. Senators Introduce Bill to Protect Aborted Babies from Medical Experimentation – Daily Citizen

In late January, pro-life senators introduced legislation to protect the human remains of aborted babies from medical experimentation for research purposes.

The bill,S. 3713, is titledProtecting Life and Integrity in Research Act of 2024.

If passed, the measure would prohibit the federal government from funding, approving, or in any way supporting research on aborted babies.

The language of S. 3713 expressly forbids medical research organization to solicit or accept aborted fetal human remains as a research donation.

Senator Cindy Hyde-Smith, chair of theSenate Pro-Life Caucus, is the bills primary sponsor. Seventeen other senators join her ascosponsors.

The law regulating human fetal tissue research is complex. Over the years, federal policy has changed significantly based on the views of each presidential administration.

Currentfederal lawpermits research on fetal human tissue if human embryos are not intentionally created or destroyed for the explicit purpose of research.

According to those statutory stipulations, it is still lawful for taxpayer dollars to fund research on aborted babies if the aborting mothers consent.

Opponents of this practice contend that taxpayers money should not be used to promote unethical research on children.

In apress release, Senator Hyde-Smith called the harvesting and trafficking of aborted babies body parts heinous and unethical.

Proponents tout the possibility of discovering medical advancements by using human fetal tissue, but recentanalysisby the Charlotte Lozier Institute calls that assumption into question.

The reportconcludesthat medical research using ethically non-controversial adult and induced pluripotent stem cells continues to advance in the pursuit of cures and treatments, while embryonic stem cells have largely fallen by the wayside, proving that science does not need to kill in order to cure.

Since 2015, the National Institutes of Healths funding for human fetal tissue research has been as high as $115 million annually. Spending for 2024 isprojectedto be approximately $61 million.

Source: National Institutes of Health

Focus on the Family, asreportedby the Daily Citizen, believes human fetal tissue research requiring the destruction of human beings is a violation of the sanctity of life.

Every human, in every condition from the single cell stage of development to natural death, is made in Gods image and possesses inestimable worth. Our human nature not our size, level of development, environment or functional capacity gives us worth and dignity as human beings. Therefore, devaluing and destroying the life of a human embryo opens the door to the devaluing and destroying any human life.

According to supporters of the legislation, the bill is endorsed by Susan B. Anthony Pro-Life America, Americans United for Life, Catholic Vote, March for Life Action, U.S. Conference of Catholic Bishops Committee on Pro-Life Activities, Students for Life Action, and Concerned Women for America LAC.

A companion measure was introduced in the U.S. House of Representatives last year,H.R. 398, but no action has been taken to advance the legislation.

The Senate bill was referred to the Senate Committee on Health, Education, Labor, and Pensions, where it awaits further action. The Daily Citizen will keep you updated on its progress.

Image from Shutterstock.

See the original post here:
U.S. Senators Introduce Bill to Protect Aborted Babies from Medical Experimentation - Daily Citizen

Embryo Patrol. Artificial Embryos Are Not Human Babies | by Karen Marie Shelton | ILLUMINATION-Curated | Jan, 2024 – Medium

Artificial Embryos Are Not Human Babies Didactic Model of Human Embryonic Development Wagner Souza Esilva Wikimedia

Artificial embryos are not human. Theyre simply a cluster of cells. To be legally human, they must meet the definition of an in vitro fertilized human ovum.

As of the end of 2023, artificial embryos couldnt be successfully implanted into mammals or humans. They couldnt lead to pregnancies, and there is no plan for that to happen in the future.

Synthetic embryos utilize stem cells groundbreakingly, sidestepping the need for sperm or eggs. Ongoing breakthroughs might eventually aid research into genetic disorders and improve babies health, including reducing the risk of problem pregnancies and miscarriages.

Artificial embryos are not related to in vitro fertilization (IVF), which can lead to a human pregnancy.

The term is misleading. These structures arent really synthetic, nor are they exactly embryos. But theyre similar. They are tiny balls of cells arising from a sperm fertilizing an egg but created from stem cells grown in the lab.

Synthetic human embryos, or SHEEFs (synthetic human entities with embryo-like features), are created from very early (actually pre-embryonic) zygotic cells called stem cells.

The stem cells are called pluripotent because they have the potential to develop into almost every cell of the body.

The lab-created embryos are not connected to a beating heart or a brain. They do include cells that would typically go on to form a version of a placenta, yolk sac, and embryo itself.

The model embryos, which resemble human versions, recreate the earliest stages of human development. They could provide a crucial window into genetic disorders and the underlying biological causes of recurrent miscarriage.

Robin Lovell-Badge, headent head of stem cell biology and developmental genetics at Francis Crick Institute in London, reported project advancements. She explained weve cultivated embryos to a specific stage just beyond what is equivalent to 14 days of development for a natural embryo.

View original post here:
Embryo Patrol. Artificial Embryos Are Not Human Babies | by Karen Marie Shelton | ILLUMINATION-Curated | Jan, 2024 - Medium

Clinical applications of stem cell-derived exosomes | Signal Transduction and Targeted Therapy – Nature.com

Kimbrel, E. A. & Lanza, R. Next-generation stem cells - ushering in a new era of cell-based therapies. Nat. Rev. Drug. Discov. 19, 463479 (2020).

Article CAS PubMed Google Scholar

Puri, M. C. & Nagy, A. Concise review: embryonic stem cells versus induced pluripotent stem cells: the game is on. Stem Cells. 30, 1014 (2012).

Article CAS PubMed Google Scholar

Ng, A. P. & Alexander, W. S. Haematopoietic stem cells: past, present and future. Cell Death Discov. 3, 17002 (2017).

Article PubMed PubMed Central Google Scholar

Naji, A. et al. Biological functions of mesenchymal stem cells and clinical implications. Cell Mol. Life Sci. 76, 33233348 (2019).

Article CAS PubMed Google Scholar

Tang, Y., Yu, P. & Cheng, L. Current progress in the derivation and therapeutic application of neural stem cells. Cell Death Dis. 8, e3108 (2017).

Article CAS PubMed PubMed Central Google Scholar

Chambers, S. E. J. et al. Current concepts on endothelial stem cells definition, location, and markers. Stem Cells Transl. Med. 10, S54S61 (2021).

Article PubMed PubMed Central Google Scholar

Hoang, D. M. et al. Stem cell-based therapy for human diseases. Signal Transduct. Target Ther. 7, 272 (2022).

Article PubMed PubMed Central Google Scholar

Zakrzewski, W., Dobrzynski, M., Szymonowicz, M. & Rybak, Z. Stem cells: past, present, and future. Stem Cell Res. Ther. 10, 68 (2019).

Article CAS PubMed PubMed Central Google Scholar

Zhang, K. & Cheng, K. Stem cell-derived exosome versus stem cell therapy. Nat. Rev. Bioeng. 1, 608609 (2023).

Article Google Scholar

Zhang, Y. et al. Exosome: A Review Of Its Classification, Isolation Techniques, Storage, Diagnostic And Targeted Therapy Applications. Int. J. Nanomed. 15, 69176934 (2020).

Article CAS Google Scholar

Vizoso, F. J. et al. Mesenchymal stem cell secretome: toward cell-free therapeutic strategies in regenerative medicine. Int. J. Mol. Sci. 18, 18521875 (2017).

Article PubMed PubMed Central Google Scholar

Ren, K. Exosomes in perspective: a potential surrogate for stem cell therapy. Odontology. 107, 271284 (2019).

Article CAS Google Scholar

Hastuti, S. et al. hUMSC vs. hUMSC-Exosome: which one is better for epilepsy? Pharmaceuticals. 15, 12471261 (2022).

Article CAS PubMed PubMed Central Google Scholar

Carr, N. J. The pathology of healing and repair. Surgery 40, 1319 (2022).

Google Scholar

Peshkova, M. et al. Targeting inflammation and regeneration: scaffolds, extracellular vesicles, and nanotechnologies as cell-free dual-target therapeutic strategies. Int. J. Mol. Sci. 23, 1379613813 (2022).

Article CAS PubMed Central Google Scholar

Wang, H., Huber, C. C. & Li, X. P. Mesenchymal and neural stem cell-derived exosomes in treating alzheimeras disease. Bioengineering 10, 253266 (2023).

Article CAS PubMed PubMed Central Google Scholar

Li, X. et al. Neural stem/progenitor cell-derived extracellular vesicles: a novel therapy for neurological diseases and beyond. MedComm 4, e214 (2023).

Article CAS PubMed Central Google Scholar

Norouzi-Barough, L., Shirian, S., Gorji, A. & Sadeghi, M. Therapeutic potential of mesenchymal stem cell-derived exosomes as a cell-free therapy approach for the treatment of skin, bone, and cartilage defects. Connect Tissue Res. 63, 8396 (2022).

Article CAS Google Scholar

Doyle, L. M. & Wang, M. Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 8, 727750 (2019).

Article CAS PubMed PubMed Central Google Scholar

Pegtel, D. M. & Gould, S. J. Exosomes. Annu. Rev. Biochem. 88, 487514 (2019).

Article CAS PubMed Google Scholar

Gurung, S., Perocheau, D., Touramanidou, L. & Baruteau, J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun. Signal. 19, 47 (2021).

Article CAS PubMed PubMed Central Google Scholar

Zhang, Y., Liu, Y., Liu, H. & Tang, W. H. Exosomes: biogenesis, biologic function and clinical potential. Cell Biosci. 9, 19 (2019).

Article PubMed PubMed Central Google Scholar

Gholami Farashah, M. S. et al. Bone marrow mesenchymal stem cells exosomes as key nanoparticles in osteogenesis and bone regeneration: specific capacity based on cell type. Mol. Biol. Rep. 49, 1220312218 (2022).

Article CAS PubMed Google Scholar

Qing, L., Chen, H., Tang, J. & Jia, X. Exosomes and their MicroRNA cargo: new players in peripheral nerve regeneration. Neurorehabilit. Neural Repair. 32, 765776 (2018).

Article Google Scholar

Kwok, Z. H., Wang, C. & Jin, Y. Extracellular vesicle transportation and uptake by recipient cells: a critical process to regulate human diseases. Processes 9, 273294 (2021).

Article CAS PubMed Google Scholar

Kimiz-Gebologlu, I. & Oncel, S. S. Exosomes: large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J. Control Release 347, 533543 (2022).

Article CAS PubMed Google Scholar

Hamzah, R. N., Alghazali, K. M., Biris, A. S. & Griffin, R. J. Exosome traceability and cell source dependence on composition and cell-cell cross talk. Int. J. Mol. Sci. 22, 53465362 (2021).

Article CAS PubMed PubMed Central Google Scholar

Li, M. et al. Exosomes from different cells: characteristics, modifications, and therapeutic applications. Eur. J. Med. Chem. 207, 112784 (2020).

Article CAS PubMed Google Scholar

Shan, X. et al. The biogenesis, biological functions, and applications of macrophage-derived exosomes. Front. Mol. Biosci. 8, 715461 (2021).

Article CAS PubMed PubMed Central Google Scholar

Wang, Y. et al. Macrophage-derived extracellular vesicles: diverse mediators of pathology and therapeutics in multiple diseases. Cell Death Dis. 11, 924 (2020).

Article CAS PubMed PubMed Central Google Scholar

Liu, J., Wu, F. & Zhou, H. Macrophage-derived exosomes in cancers: biogenesis, functions and therapeutic applications. Immunol. Lett. 227, 102108 (2020).

Article CAS Google Scholar

Elashiry, M., Elsayed, R. & Cutler, C. W. Exogenous and endogenous dendritic cell-derived exosomes: lessons learned for immunotherapy and disease pathogenesis. Cells. 11, 115136 (2021).

Article PubMed PubMed Central Google Scholar

Pitt, J. M. et al. Dendritic cell-derived exosomes for cancer therapy. J Clin Investig. 126, 12241232 (2016).

Article PubMed Central Google Scholar

Kok, V. C. & Yu, C. C. Cancer-derived exosomes: their role in cancer biology and biomarker development. Int. J. Nanomed. 15, 80198036 (2020).

Article CAS Google Scholar

Naseri, M. et al. Tumor-derived exosomes: the next generation of promising cell-free vaccines in cancer immunotherapy. Oncoimmunology 9, 1779991 (2020).

Article PubMed PubMed Central Google Scholar

Kluszczynska, K. et al. Methods for the determination of the purity of exosomes. Curr. Pharm. Des. 25, 44644485 (2019).

Article CAS PubMed Google Scholar

Zhou, B. et al. Application of exosomes as liquid biopsy in clinical diagnosis. Signal Transduct Target Ther. 5, 144 (2020).

Article CAS PubMed Central Google Scholar

Boukouris, S. & Mathivanan, S. Exosomes in bodily fluids are a highly stable resource of disease biomarkers. Proteomics Clin. Appl. 9, 358367 (2015).

Article CAS PubMed PubMed Central Google Scholar

Gurunathan, S. et al. Review of the isolation, characterization, biological function, and multifarious therapeutic approaches of exosomes. Cells. 8, 307342 (2019).

Article CAS PubMed Central Google Scholar

Bei, H. P. et al. Bone-a-Petite: engineering exosomes towards Bone, osteochondral, and cartilage repair. Small. 17, e2101741 (2021).

Article PubMed Google Scholar

Yang, X. X., Sun, C., Wang, L. & Guo, X. L. New insight into isolation, identification techniques and medical applications of exosomes. J. Control Release 308, 119129 (2019).

Article CAS PubMed Google Scholar

Kalluri, R. & LeBleu, V. S. The biology, function, and biomedical applications of exosomes. Science. 367, 640654 (2020).

Article Google Scholar

Li, X. et al. Challenges and opportunities in exosome research-Perspectives from biology, engineering, and cancer therapy. APL Bioeng. 3, 011503 (2019).

Article PubMed PubMed Central Google Scholar

Khayambashi, P. et al. Hydrogel encapsulation of mesenchymal stem cells and their derived exosomes for tissue engineering. Int. J. Mol. Sci. 22, 684698 (2021).

Article CAS PubMed PubMed Central Google Scholar

Hussen, B. M. et al. Strategies to overcome the main challenges of the use of exosomes as drug carrier for cancer therapy. Cancer Cell Int. 22, 323 (2022).

Article PubMed PubMed Central Google Scholar

Shao, J., Zaro, J. & Shen, Y. Advances in exosome-based drug delivery and tumor targeting: from tissue distribution to intracellular fate. Int. J. Nanomed. 15, 93559371 (2020).

Article CAS Google Scholar

Tian, Y. et al. A doxorubicin delivery platform using engineered natural membrane vesicle exosomes for targeted tumor therapy. Biomaterials 35, 23832390 (2014).

Article CAS Google Scholar

Kim, M. S. et al. Development of exosome-encapsulated paclitaxel to overcome MDR in cancer cells. Nanomedicine 12, 655664 (2016).

Article CAS PubMed Google Scholar

Ohno, S. et al. Systemically injected exosomes targeted to EGFR deliver antitumor microRNA to breast cancer cells. Mol. Ther. 21, 185191 (2013).

Article CAS PubMed Google Scholar

Alvarez-Erviti, L. et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat. Biotechnol. 29, 341345 (2011).

Article CAS PubMed Google Scholar

More:
Clinical applications of stem cell-derived exosomes | Signal Transduction and Targeted Therapy - Nature.com

Developing in-vivo chimeric lungs with pluripotent stem cells – Drug Target Review

Reverse-blastocyst complementation elucidates the conditions required to form lungs in rat-mouse chimeric models.

Researchers from the Nara Institute of Science and Technology (NAIST) have used the reverse-blastocyst complementation (rBC) method to understand the conditions required to form lungs in rat-mouse chimeric models. They also used the tetraploid-based organ complementation (TOC) method to create a rat-derived lung in their mouse model.

Chronic obstructive pulmonary disease (COPD) is the third leading cause of global deaths. The pathogenesis of COPD is based on the innate and adaptive inflammatory immune response to the inhalation of toxic particles and gases. Although tobacco smoking is the primary cause of this inhalation injury, many other environmental and occupational exposures contribute to the pathology of COPD.1

Lung transplantation is the only viable treatment option, yet finding suitable lung donors is difficult. However, regenerative medicine is advancing the development of lungs from pluripotent stem cells (PSCs) using interspecies animal models. Current expectations for realisation of the promise of PSCs are at the highest they have ever been. However, there are many challenges that need to be addressed in order to bring PSC technology within the grasp of many more patients. Three particular challenges are: tumorigenicity, immunogenicity, and heterogeneity.2

PSCs and embryonic stem cells (ESCs) from one species can be injected into blastocytes, the cluster of diving cells made by a fertilised egg, in a biological technique named blastocyst complementation to create interspecies chimeric animals. This has enabled successful regeneration of the heart, pancreas and kidney in rat-mouse chimeras, but functional lung formation remains to be achieved successfully in vitro due to the complex three-dimensional (3D) structures and multiple cell types needed. This has warranted more research into the viable conditions required to generate PSC-derived organs.

Fibroblast growth factors (FGFs)are polypeptides with various biological activities bothin vivoandin vitro.3 The fibroblast growth factor 10 (Fgf10) and its interaction with the Fgf receptor isoform IIIb (Fgfr2b) in the lungs are essential for lung development. The rBC method in the new study involved injecting mutant ESCs which fail to show lung formation into wild-type (WT) embryos. This allowed for efficient detection of mutant PSCs in the recipient tissue, aiding the determination of the conditions necessary for successful lung formation in the organ-deficient animal.

The team, led by Dr Shunsuke Yuri and Dr Ayako Isotani, discovered that WT ESCs provide uniform contributions across target and non-target organs in the chimeras. This helped to ascertain that a particular number of WT or normal cells are required to overcome the lung development failure in Fgf10-deficient or Fgfr2b-deficient animals.

Having this understanding enabled them to produce rat-derived lungs in the Fgfr2b-deficient mouse embryos with the TOC method, without the requirement of producing a mutant mouse line. Dr Yuri said: Interestingly, we found that the rat epithelial cells conserved intrinsic species-specific timing in the interspecies model, resulting in an underdeveloped lung. Consequently, these lungs remained nonfunctional post-birth.

The studys findings identified the factors required for the successful generation of functional lungs in rat-mouse interspecies chimeras, as well as the issues to overcome. Dr Yuri concluded: We believe that our study makes an important contribution to the literature by presenting a faster and more efficient method of exploring blastocyst complementation.

These novel results can significantly advance the progress toward developingin-vivochimeric lungs for the purpose of transplantation, which could transform the practical application of regenerative medicine.

This study was published in Development.

1 Hogg JC, Timens W. The Pathology of Chronic Obstructive Pulmonary Disease. Annual Review of Pathology: Mechanisms of Disease. 2008 October 27 [2024 January 5]; 4:435-459. Available from: https://doi.org/10.1146/annurev.pathol.4.110807.092145

2 Yamanaka S. Pluripotent Stem Cell-Based Cell Therapy Promise and Challenges. Cell Stem Cell. 2020 October 1 [2024 January 5]; 27(4):523-31. Available from: https://doi.org/10.1016/j.stem.2020.09.014

3 Birnbaum D, Coulier F, Emoto H, Itoh N, Mattei MG, Tagashira S. Structure and Expression of Human Fibroblast Growth Factor-10*. Journal of Biological Chemistry. 1997 September [2024 January 2024]; 272(37):23191-4. Available from: https://doi.org/10.1074/jbc.272.37.23191

View original post here:
Developing in-vivo chimeric lungs with pluripotent stem cells - Drug Target Review

No, Rep. Steve Scalise Didn’t Vote Against Stem Cell Research From Which He Is Now Benefiting – Yahoo News

A long-dormant medical controversy was revived last week following an announcement from House Majority Leader Steve Scalise. On January 5, his office released a statement indicating that he was undergoing a stem cell transplant as part of his previously announced treatment for multiple myeloma. Hearkening back to the stem cell controversies of the early 2000s, a number of posts emerged onlineincluding a viral Reddit thread and tweet with 54,000 likes and almost 10,000 retweetsaccusing the congressman of hypocrisy for receiving a treatment that he allegedly fought against.

The treatment Scalise is receiving has no relation to the embryonic stem cell research often opposed by pro-life Americans, however, and the congressman has never voted to restrict research into the form of treatment he is receiving.

Scalise first announced his diagnosis of multiple myelomaa rare blood cancerin late August 2023, telling reporters a month later that his body had responded well to his first round of treatment. The good news is the cancer has dropped dramatically because of the success of the chemotherapy, he said in September. The next step for Scalise, as mentioned above, is an autologous stem cell transplant. [Rep. Scalise] is currently undergoing the transplant process, marking a significant milestone in his battle against cancer, his offices January 5 statement read. Once the procedure is completed, he will be recovering under the supervision of his medical team and will work remotely until returning to Washington next month. Scalise is receiving treatment in his home state of Louisiana.

Because multiple myeloma attacks a patients bone marrowan essential tissue for the bodys production of blood cellsstem cell transplants are often used to help replace marrow damaged by the cancer with new and healthy marrow. In a typical autologous stem cell treatment, the kind which Scalise is receiving, a patients own hematopoietic stem cells are extracted and frozen multiple weeks before treatment. These cells used to be extracted from the bone marrow itself, but today most patients are given a growth factor that allows for stem cells to be taken directly from the blood. The patient is then given intensive chemotherapy, often in a single large dose, before receiving a transfusion of his or her own healthy stem cells. It then takes two to three weeks for the transfused stem cells to restore the functionality of the bone marrow, during which patients can be substantially immunocompromised because of their bodies inability to produce the white blood cells necessary for proper immune function.

Unlike embryonic stem cells, which are harvested from early stage human embryos and can take the form of any cell in the body, the hematopoietic stem cells used in the treatment of multiple myeloma are extracted from a patients own body or from a voluntary donor and can develop into only a limited range of blood cells. These are not the type of stem cells that are in an embryo that can become anything, Dr. Marc Braunstein, a hematology and stem cell transplant expert at NYU Langone Health, told The Dispatch Fact Check. These are slightly differentiated stem cells that are destined to become blood cells, but not anything else.

Traditional stem cell therapies are widely accepted and utilized in modern medical practice, unlike the embryonic stem cell research that reached a point of national controversy in the mid-2000s. We can debate the ethics of using embryonic stem cells, Braunstein said, but I think in this case were not talking about that at all. According to Braunstein, even patients who are practicing Jehovahs Witnessa religious group that typically rejects the use of blood transfusionsare often not opposed to autologous stem cell treatments. For those individuals who may be leery about the use of embryonic stem cells, I dont think they would be as concerned with the use of adult hematopoietic stem cells, Braunstein said.

Furthermore, Scalise has not taken any notable votes against stem cell researchembryonic or non-embryonic. Two notable bills intended to advance embryonic stem cell research, the Stem Cell Research Enhancement Act of 2005 and Stem Cell Research Enhancement Act of 2007, passed both the House and Senate, but both were vetoed by then President George W. Bush. These votes occurred prior to Scalise assuming office in May 2008, however, and very little legislative activity involving embryonic stem cell research has happened since.

In September 2020, Scalise co-signed a letter by Mississippi Sen. Roger Wicker calling for an end to taxpayer funded embryonic stem cell research at the National Institutes of Health, but the letter expressed no opposition to non-embryonic stem cell research or treatment. In fact, Scalise voted in favor of the Stem Cell Therapeutic and Research Reauthorization Act of 2010, Stem Cell Therapeutic and Research Reauthorization Act of 2015, and TRANSPLANT Act of 2021, all of which reauthorized a program intended to support patients in need of stem cell transplants.

Asked by The Dispatch Fact Check whether they believed allegations of hypocrisy were unfair, Scalises office declined to comment further, instead saying that the statement on his treatment spoke for itself.

If you have a claim you would like to see us fact check, please send us an email at factcheck@thedispatch.com. If you would like to suggest a correction to this piece or any other Dispatch article, please email corrections@thedispatch.com.

Read more at The Dispatch

The Dispatch is a new digital media company providing engaged citizens with fact-based reporting and commentary, informed by conservative principles. Sign up for free.

The rest is here:
No, Rep. Steve Scalise Didn't Vote Against Stem Cell Research From Which He Is Now Benefiting - Yahoo News

Embryonic-stem-cell-derived mesenchymal stem cells relieve experimental contact urticaria by regulating the functions … – Nature.com

Animals

All animal experimental procedures were approved by the Dong-A University Medical School Institutional Animal Care and Use Committee (Approval No. DIACUC-21-11) at 22.03.2021. Female BALB/c mice that were 78weeks old were purchased from Orient Bio Inc. (Gyeonggi-do, korea). The classification of experimental groups involves the random assignment of mice of the same age and within a 1g difference in body weight after the acclimatization process. The mice are then housed in a pathogen-free facility at Dong-A University (Busan, Korea). Mice were maintained at Dong-A University facility at 22C1C room temperature, 4060% humidity, on a 12h lightdark cycle (7a.m. to 7p.m.), and given food and water freely, according to institutional guidelines. All experiments were performed under inhalation anesthesia with isoflurane, and mice were euthanized by CO2 inhalation at the end of the experiment. This study adhered to the guidelines set forth by the laboratory animal ethics committee of Dong-A University and the ARRIVE guidelines. To ensure statistical significance, 5 or more mice per group were used, and all experimental protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of Dong-A University. Inhalational anesthesia using isoflurane was used to induce anesthesia when sacrificing all experimental animals.

Contact urticaria mouse model was induced according to a previously reported method31,32. Mice were initially sensitized by applying 100l of a TMA (trimellitic anhydride; 500mg/ml, Alfa Aesar, Ward Hill, MA, USA) in acetone/olive oil (4:1, v/v) on the shaved hind flank. This sensitization process is essential for inducing an immune response to TMA. Secondary sensitizations were performed on the hind flank to reinforce the immune response. On the 7th and 10th days after the first sensitization, mice sensitized 50l of a TMA solution (250mg/ml) in acetone/olive oil (4:1, v/v). On day 13 after the initial sensitization, contact urticaria (CU) was induced by challenging the ears with 25l of a TMA solution (100mg/ml) dissolved in acetone/olive oil (4:1, v/v). The disease symptoms were assessed by measuring ear thickness, itching, and lesions on the skin. Particularly, skin lesions were evaluated by determining the ratio of the affected area, indicating erythema and edema on a 3 cm2 area of the dorsal skin. The symptom evaluation of experimental animals was evaluated based on all animals without exclusion criteria. For histological analysis, H&E and mast cell staining were conducted. Cellular and molecular analysis involved the use of flow cytometry to assess immune cell activity in mouse lymphoid organs, along with genetic analysis of the lesions. Experimental animals of 10 mice per group were performed by blindly selecting 56 mice for efficient handling of animal-derived flow cytometric and genetic analysis. The corresponding author and one of the first authors (S.Y. Hyun) were aware of the group assignment, and the symptoms, flow cytometry, and other molecular analysis results were evaluated together by multiple blinded co-authors.

For the purposes of in vitro experiments, we used the nave CD3+ T cell isolation Kit (Miltenyi Biotec, Bergisch-Gladbach, Germany) to enrich nave CD3+ cells from the spleens of BALB/c mice (6weeks old). All steps were conducted strictly following the manufacturers protocol.

M-MSCs used in this study was provided Mirae Cell Bio (Seoul, Korea). M-MSCs differentiated from H9 hESCs30 were maintained in EGM2-MV medium (Lonza, San Diego, CA, USA) containing supplement Mix (promocell, Heidelberg, Germany) and 50 ug/ml Gentamicin (Gibco, NewYork, USA) in a humidified atmosphere containing 5% CO2 at 37, as previously described33. M-MSCs at less than ten passages were used for in vitro cell culture and in vivo animal experiments. Bone marrow-derived MSCs (BM-MSCs) were maintained in MSCBM medium (Lonza, Basel, Switzerland) containing supplement kit (Lonza) in a humidified incubator at 5% CO2 at 37. Bone marrow-derived mast cells (BMMCs) derived from BALB/c mice were cultured in RPMI 1640 medium containing 2mM L-glutamine, 0.1mM nonessential amino acids, antibiotics, 10% fetal bovine serum (FBS), and IL-3 (10ng/ml; PeproTech Inc., Rocky hill, NJ, USA). After 4weeks,>98% of the cells were verified as BMMCs, as previously described34. Mouse splenic T cells were presorted by CD3 mAb-microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany) followed by the manufacturer's method. For T cell polarization, splenic nave CD3+ T cells were cultured onto a 24-well plate coated with 1g/ml of anti-CD3 (eBioscience, San Diego, CA, USA) in complete RPMI 1640 medium and supplemented with TH1 reagents [IL-2 (20ng/ml, PeproTech Inc.), IL-12 (20ng/ml, PeproTech Inc.), and anti-IL-4 (10g/ml, Bio X cell, West Lebanon, NH, USA)] or TH2 reagents [IL-2 (20ng/ml, PeproTech Inc.), IL-4 (100ng/ml, PeproTech Inc.), and anti-IFN- (10g/ml, Bio X cell)]. After 48h, the cells were co-cultured with M-MSCs for 24h under polarized conditions. To confirm the regulation of immune cells by M-MSCs in vitro, BMMCs (5.0105 cells/well), splenic T cells (5.0105 cells/well), or polarizing splenic T cells (5.0105 cells/well) were co-cultured with the indicated ratio of M-MSCs for 24h. Co-cultured splenic T cells were identified by flow cytometry analysis. The analysis of polarized T cells was assessed by distinguishing them using the gating strategy as depicted in Supplementary Fig.S1. The ratio of degranulation of BMMCs was analyzed by -hexosaminidase secretion.

After the primary sensitization of the contact urticaria model, M-MSCs were injected subcutaneously into the ear as a single administration on the 10th days or twice on the 10th and 12th days. A primary disease improvement evaluation was performed through single administration or two administrations, and an appropriate cell administration group for disease efficacy was selected. The administration of an equal amount of BM-MSCs and oral administration of cetirizine (50mg/kg) (Sigma-Aldrich, St. Louis, MO, USA) were used as positive controls. To deplete TGF-, BALB/c mice were intraperitoneally injected with 300g of anti-TGF- mAb (1D11.16.8, Bio X cell) or an isotype-matched control mAb (Bio X cell) twice on days 0 and 3 of M-MSCs administration.

Single-cell suspensions were isolated from the spleen, cervical lymph node (cLN), and ear. Ear tissues were isolated into single cells using the gentleMACS dissociator (Miltenyi Biotec) followed by the manufacturers method. For the detection of intracellular cytokines, the isolated cells were stimulated with PMA (50ng/ml; Sigma-Aldrich), ionomycin (500ng/ml; Sigma-Aldrich), and brefeldin A (3g/ml; eBioscience) for 4h before analysis and a fixation/permeabilization kit were from eBioscience. Before cell surface markers were stained, Fc receptors were blocked with anti-CD16 and anti-CD32 mAbs (2.4G2, BD Biosciences), and conjugated and dead cells were excluded by analysis on the basis of forward and side light scatter parameters and staining with a Zombie NIR Fixable Viability Kit (Biolegend, San Diego, CA, USA). The antibodies against proteins were as follows: Antibodies against CD3 (17A2) and CD8a (53-6.7) were obtained from BioLegend. Antibodies for CD4 (RM4-5), IFN- ((XMG1.2), IL-4 (11B11) were obtained from eBioscience. Antibodies for CD3 (17A2), CD45 (30-F11), and CD127 (A7R34) were obtained from BioLegend. The cells were then analyzed with a NovoCyte flow cytometer (Agilent) and FlowJo version 10 software (Tree Star, Ashland, OR, USA).

BMMCs (5.0105 cells/well) co-cultured with M-MSCs (0.5 to 2106 cells/well) for 24h were sensitized for 4h with Monoclonal dinitrophenol (DNP)-specific IgE (100ng/ml; Sigma). The IgE-primed BMMCs were then stimulated with 50ng/ml of DNP-human serum albumin (DNP-HSA, Sigma-Aldrich) in Tyrode-BSA buffer (20mM Hepes (pH 7.4), 135mM NaCl, 5mM KCl, 1.8mM CaCl2, 1mM MgCl2, 5.6mM glucose, and 0.1% BSA) for 15min in the presence or absence of the M-MSCs.

Degranulation was determined by measuring the release of the granule marker -hexosaminidase as previously described35. The degree of degranulation of BMMCs was expressed as the % of the activity of -hexosaminidase secreted out of the cells compared to the total activity of -hexosaminidase.

M-MSCs were co-cultured with splenic T cells or BMMCs for 24h and then effector cells were removed. M-MSCs were rinsed with PBS and left on ice for 5min to stop the reaction. Total RNA was extracted using AccuPrep Universal RNA Extraction Kit (Bioneer, Daejeon, Korea), and cDNA was synthesized using AccuPower CycleScript RT PreMix (Bioneer) according to the manufacturers instructions. The PCR reaction was amplified using AccuPower PCR PreMix (Bioneer) and PCR was performed at 95 for 2min, 95 for 20s, 58 for 40s, 72 for 30s, 72 for 5min for 30 cycles. Primers used as follow: human Hgf (forward 5-TCCATGATACCACACGAACACAGC-3, reverse 5-TGCACAGTACTCCCAGCGGGTGTG-3); human Ido1 (forward 5-TTTGCTAAAGGCGCTGTTGG-3, reverse 5-CCTTCATACACCAGACCGTCTGA-3); human Pdl1 (forward 5-TATGGTGGTGCCGACTACAA-3, reverse 5-TGCTTGTCCAGATGACTTCG-3); human Il10 (forward 5-AGACATCAGGGTGGCGACTCTAT-3, reverse 5-GGCTCCCTGGTTTCTCTTCCTAAG-3); human Pge2 (forward 5-ACCATCTACCCCTTCCTTT-3, reverse 5-CCGCTTCCCAGAGGATCT-3); human Tgfb (forward 5-GGGACTATCCACCTGCAAGA -3, reverse 5-CCTCTTGGCGTAGTAGTCG-3); human Gapdh (forward 5-ACCACAGTCCATGCCATCAC-3, reverse 5-TCCACCACCCTGTTGCTGTA-3). Snap-frozen disease-inducing mouse ear tissues were ground to powder. Total RNA isolation and PCR reaction were performed in the same manner as above. Real-time PCR was performed Thermal Cycler Dice Real Time System III TP950 (Takara, Shiga-ken, Japan). Primers used as follow: mouse Il4 (forward 5-ACAGGAGAAGGGACGCCAT-3, reverse 5-GAAGCCCTACAGACGAGCTCA-3); mouse Il6 (forward 5-GAGGATACCACTCCCAACAGACC-3, reverse 5-AAGTGCATCATCGTTGTTCATACA-3); mouse Ifng (forward 5-CAGCAACAGCAAGGCGAAAAAGG-3, reverse 5-TTTCCGCTTCCTGAGGCTGGAT-3); mouse Tnfa (forward 5-AGTGACAAGCCTGTAGCCCACGT -3, reverse 5-CCATCGGCTGGCACCACTAGTT-3); mouse Gapdh (forward 5-CATCACTGCCACCCAGAAGACTG-3, reverse 5-ATGCCAGTGAGCTTCCCGTTCAG-3);

After the induction of contact urticaria in mice, their ear tissues were fixed in 4% paraformaldehyde in phosphate-buffered saline for 24h and then embedded in paraffin. The tissues were dehydrated in a graded ethanol series (70 to 100%), rinsed three times with xylene for 3min each, and then embedded in paraffin. Sections of paraffin-embedded tissues, with a thickness of 6m, were prepared and stained with hematoxylin (Sigma-Aldrich) and eosin (Sigma-Aldrich) to compare and analyze the degree of cell invasion and epidermal thickness in the tissue. Additionally, sections of tissues with a thickness of 6m were stained with a 1% toluidine blue (Sigma-Aldrich) solution to assess the number of infiltrating mast cells and the degree of degranulation.

The in vitro experiment was repeated three independent times, and the animal experiment was based on five or more animals per group, and if the results of the first experiment were insufficient, the significance was evaluated within a total of 10 animals in the group. The data are presented as the meanstandard error (SEM) from three or more independent experiments for in vitro experiments. Statistical analysis was done by unpaired Student's t-test. One-way analysis of variance (ANOVA) with Tukey's post hoc test was performed for multiple comparisons. Statistical significance (*P<0.05 and **P<0.01) was determined with Prism version 7.0 (GraphPad, San Diego, CA).

This study was approved by the Institutional Animal Care and Use Committee (IACUC) of Dong-A University(DIACUC-21-11). All animal experiments were performed in accordance with the guidelines and regulationsof the institutional guidelines.

Go here to read the rest:
Embryonic-stem-cell-derived mesenchymal stem cells relieve experimental contact urticaria by regulating the functions ... - Nature.com