Stem Cells: Getting to the heart of cardiovascular evolution in humans

Differences in the response of cardiomyocytes to oxygen deprivation in humans and chimpanzees may explain why humans are more prone to certain heart diseases.
  1. Alex Pollen  Is a corresponding author
  2. Bryan J Pavlovic
  1. University of California, San Francisco, United States

Cardiovascular disease is one of the leading causes of death in both humans and captive chimpanzees. However, despite having similar heart anatomies, humans and chimpanzees are prone to different types of cardiovascular disease. In humans, a build-up of fatty materials in the arteries (a condition called atherosclerosis) can restrict blood flow to the heart, which can cause myocardial ischemia. On the other hand, great apes, including chimpanzees, are more likely to suffer heart disease from myocardial fibrosis (Lowenstine et al., 2016). This condition, which rarely leads to heart disease in humans, involves increased deposition of collagen in heart tissue (Lammey et al., 2008).

Both genetic and environmental factors could be responsible for these differences. Interestingly, cholesterol is a major risk factor for atherosclerosis in humans, yet captive chimpanzees manage to avoid this condition despite having higher cholesterol levels than humans (Varki et al., 2009). However, a lack of appropriate model systems has made it difficult to investigate how genetic differences in disease susceptibility evolved between these two species.

Recent studies have shown that cardiomyocytes (heart muscle cells derived from stem cells) recapitulate many of the features of normal human and chimpanzee hearts, although there are important differences in metabolism and maturation state (Pavlovic et al., 2018). Since it is possible to grow human and chimpanzee cardiomyocytes under common environmental conditions, this provides an opportunity to study differences in the genetics of the two systems (Gallego Romero et al., 2015). Now, in eLife, Michelle Ward and Yoav Gilad from the University of Chicago report how cardiomyocytes can be used to compare the genomic consequences of myocardial ischemia in humans and chimpanzees (Ward and Gilad, 2019).

A characteristic symptom of myocardial ischemia is oxygen deprivation, also known as hypoxia, which is caused by a reduction in the flow of blood to the heart. To simulate hypoxia, Ward and Gilad first cultured cardiomyocytes in normal oxygen levels, then exposed them to a ten-fold decrease in oxygen for six hours, before returning them to normal oxygen levels. Depriving heart tissue of oxygen typically leads to increased production of reactive oxygen species (such as peroxides) that can cause DNA damage and lipid degradation. Hypoxia induced similar effects in both human and chimpanzee cardiomyocytes, suggesting that such experiments can recapitulate the effects of myocardial ischemia.

RNA sequencing revealed that nearly one third of the genes that are expressed in cardiomyocytes respond to hypoxia. Moreover, 75% of these responded in the same way in both species, suggesting that hypoxia has a widespread and largely conserved effect on gene expression. In contrast, previous work has shown that the immune responses in humans and chimpanzees are quite different (Barreiro et al., 2010).

Next, Ward and Gilad investigated how patterns of gene expression were influenced by hypoxia within these two species. Despite the similarities revealed in the RNA sequencing experiments, several hundred genes still differed in their responses. One possible explanation for these differences is that hypoxia-related transcription factors bind to response genes to varying degrees. Consistent with this possibility, hypoxia-related factors were shown to frequently bind to, or near to, conserved response genes, but not to genes with chimpanzee-specific responses. This suggests that changes in transcription factor binding, and possibly changes to the sites they bind to, may be responsible for the several hundred genes that respond differently to hypoxia in the two species.

Another notable difference is the response of a protein called RASD1, which is upregulated during hypoxia in human cardiomyocytes but not in chimpanzee cardiomyocytes. Interestingly, this gene is also upregulated in patients with myocardial ischemia and other diseases related to atherosclerosis. RASD1 could therefore have a role in regulating hypoxia-induced tissue damage in humans.

Ward and Gilad then explored the role of genes called eQTL genes (where eQTL is short for expression quantitative trait loci). Genetic variation within these genes can explain some of the disparity in mRNA levels among individuals in a population. Intersecting eQTL genes with genetic association studies has become a popular strategy for identifying disease-risk genes (Albert and Kruglyak, 2015). However, several recent lines of evidence suggest that eQTL genes may reflect mostly neutral genetic variation and are not therefore associated with any specific diseases (Jasinska et al., 2017; Tung et al., 2015).

Ward and Gilad found that there was a significant overlap between the conserved hypoxia response genes and the genes that are upregulated in patients with myocardial ischemia. This overlap indicates that the response genes are relevant to disease: however, the overlap between these genes and eQTL genes was lower than expected (Figure 1). Therefore, identifying the genetic variants and genes that influence disease-relevant responses, such as hypoxia, is likely to be more informative than traditional baseline eQTL studies. This means that stem cell models, such as those studied by Ward and Gilad, could be used more generally to study how common variation and evolved genetic differences influence other disease-relevant responses.

Using stem cell models to explore the genetic effects that influence susceptibility to cardiovascular disease.

Ward and Gilad explored the effects of hypoxia (that is, oxygen deprivation) on cardiomyocytes (heart cells derived from stem cells) from humans and chimpanzees. They found that 75% of the genes that respond to hypoxia respond in the same way in both species (third panel). They also observed a large overlap between these conserved response genes and genes that are upregulated in patients with ischemia (fourth panel; left), but little overlap with eQTL genes (right).

References

    1. Lammey ML
    2. Baskin GB
    3. Gigliotti AP
    4. Lee DR
    5. Ely JJ
    6. Sleeper MM
    (2008)
    Interstitial myocardial fibrosis in a captive chimpanzee (Pan troglodytes) population
    Comparative Medicine 58:389–394.

Article and author information

Author details

  1. Alex Pollen

    Alex Pollen is in the Department of Neurology, and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States

    For correspondence
    Alex.Pollen@ucsf.edu
    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0003-3263-8634
  2. Bryan J Pavlovic

    Bryan J Pavlovic is in the Department of Neurology, and The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, United States

    Competing interests
    No competing interests declared
    ORCID icon "This ORCID iD identifies the author of this article:" 0000-0002-7751-5315

Publication history

  1. Version of Record published: May 28, 2019 (version 1)

Copyright

© 2019, Pollen and Pavlovic

This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.

Metrics

  • 1,186
    views
  • 99
    downloads
  • 2
    citations

Views, downloads and citations are aggregated across all versions of this paper published by eLife.

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Alex Pollen
  2. Bryan J Pavlovic
(2019)
Stem Cells: Getting to the heart of cardiovascular evolution in humans
eLife 8:e47807.
https://doi.org/10.7554/eLife.47807

Further reading

    1. Evolutionary Biology
    Robert Horvath, Nikolaos Minadakis ... Anne C Roulin
    Research Article

    Understanding how plants adapt to changing environments and the potential contribution of transposable elements (TEs) to this process is a key question in evolutionary genomics. While TEs have recently been put forward as active players in the context of adaptation, few studies have thoroughly investigated their precise role in plant evolution. Here, we used the wild Mediterranean grass Brachypodium distachyon as a model species to identify and quantify the forces acting on TEs during the adaptation of this species to various conditions, across its entire geographic range. Using sequencing data from more than 320 natural B. distachyon accessions and a suite of population genomics approaches, we reveal that putatively adaptive TE polymorphisms are rare in wild B. distachyon populations. After accounting for changes in past TE activity, we show that only a small proportion of TE polymorphisms evolved neutrally (<10%), while the vast majority of them are under moderate purifying selection regardless of their distance to genes. TE polymorphisms should not be ignored when conducting evolutionary studies, as they can be linked to adaptation. However, our study clearly shows that while they have a large potential to cause phenotypic variation in B. distachyon, they are not favored during evolution and adaptation over other types of mutations (such as point mutations) in this species.

    1. Evolutionary Biology
    Zhiliang Zhang, Zhifei Zhang ... Guoxiang Li
    Research Article

    Biologically-controlled mineralization producing organic-inorganic composites (hard skeletons) by metazoan biomineralizers has been an evolutionary innovation since the earliest Cambrian. Among them, linguliform brachiopods are one of the key invertebrates that secrete calcium phosphate minerals to build their shells. One of the most distinct shell structures is the organo-phosphatic cylindrical column exclusive to phosphatic-shelled brachiopods, including both crown and stem groups. However, the complexity, diversity, and biomineralization processes of these microscopic columns are far from clear in brachiopod ancestors. Here, exquisitely well-preserved columnar shell ultrastructures are reported for the first time in the earliest eoobolids Latusobolus xiaoyangbaensis gen. et sp. nov. and Eoobolus acutulus sp. nov. from the Cambrian Series 2 Shuijingtuo Formation of South China. The hierarchical shell architectures, epithelial cell moulds, and the shape and size of cylindrical columns are scrutinised in these new species. Their calcium phosphate-based biomineralized shells are mainly composed of stacked sandwich columnar units. The secretion and construction of the stacked sandwich model of columnar architecture, which played a significant role in the evolution of linguliforms, is highly biologically controlled and organic-matrix mediated. Furthermore, a continuous transformation of anatomic features resulting from the growth of diverse columnar shells is revealed between Eoobolidae, Lingulellotretidae, and Acrotretida, shedding new light on the evolutionary growth and adaptive innovation of biomineralized columnar architecture among early phosphatic-shelled brachiopods during the Cambrian explosion.