101
|
Schneider E, El Hajj N, Richter S, Roche-Santiago J, Nanda I, Schempp W, Riederer P, Navarro B, Bontrop RE, Kondova I, Scholz CJ, Haaf T. Widespread differences in cortex DNA methylation of the "language gene" CNTNAP2 between humans and chimpanzees. Epigenetics 2014; 9:533-45. [PMID: 24434791 PMCID: PMC4121364 DOI: 10.4161/epi.27689] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CNTNAP2, one of the largest genes in the human genome, has been linked to human-specific language abilities and neurodevelopmental disorders. Our hypothesis is that epigenetic rather than genetic changes have accelerated the evolution of the human brain. To compare the cortex DNA methylation patterns of human and chimpanzee CNTNAP2 at ultra-high resolution, we combined methylated DNA immunoprecipitation (MeDIP) with NimbleGen tiling arrays for the orthologous gene and flanking sequences. Approximately 1.59 Mb of the 2.51 Mb target region could be aligned and analyzed with a customized algorithm in both species. More than one fifth (0.34 Mb) of the analyzed sequence throughout the entire gene displayed significant methylation differences between six human and five chimpanzee cortices. One of the most striking interspecies differences with 28% methylation in human and 59% in chimpanzee cortex (by bisulfite pyrosequencing) lies in a region 300 bp upstream of human SNP rs7794745 which has been associated with autism and parent-of-origin effects. Quantitative real-time RT PCR revealed that the protein-coding splice variant CNTNAP2-201 is 1.6-fold upregulated in human cortex, compared with the chimpanzee. Transcripts CNTNAP2-001, -002, and -003 did not show skewed allelic expression, which argues against CNTNAP2 imprinting, at least in adult human brain. Collectively, our results suggest widespread cortex DNA methylation changes in CNTNAP2 since the human-chimpanzee split, supporting a role for CNTNAP2 fine-regulation in human-specific language and communication traits.
Collapse
Affiliation(s)
- Eberhard Schneider
- Institute for Human Genetics; Julius Maximilian University; Würzburg, Germany
| | - Nady El Hajj
- Institute for Human Genetics; Julius Maximilian University; Würzburg, Germany
| | - Steven Richter
- Institute for Human Genetics; Julius Maximilian University; Würzburg, Germany
| | | | - Indrajit Nanda
- Institute for Human Genetics; Julius Maximilian University; Würzburg, Germany
| | - Werner Schempp
- Institute for Human Genetics; University of Freiburg; Freiburg, Germany
| | - Peter Riederer
- Clinical Neurochemistry Laboratory; Department of Psychiatry; University Hospital; Würzburg, Germany
| | - Bianca Navarro
- Institute of Legal Medicine; University Medical Center; Mainz, Germany
| | | | - Ivanela Kondova
- Biomedical Primate Research Center; Rijswijk, The Netherlands
| | - Claus Jürgen Scholz
- Laboratory for Microarray Applications; IZKF; Julius Maximilians University; Würzburg, Germany
| | - Thomas Haaf
- Institute for Human Genetics; Julius Maximilian University; Würzburg, Germany
| |
Collapse
|
102
|
Muntané G, Horvath JE, Hof PR, Ely JJ, Hopkins WD, Raghanti MA, Lewandowski AH, Wray GA, Sherwood CC. Analysis of synaptic gene expression in the neocortex of primates reveals evolutionary changes in glutamatergic neurotransmission. ACTA ACUST UNITED AC 2014; 25:1596-607. [PMID: 24408959 DOI: 10.1093/cercor/bht354] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Increased relative brain size characterizes the evolution of primates, suggesting that enhanced cognition plays an important part in the behavioral adaptations of this mammalian order. In addition to changes in brain anatomy, cognition can also be regulated by molecular changes that alter synaptic function, but little is known about modifications of synapses in primate brain evolution. The aim of the current study was to investigate the expression patterns and evolution of 20 synaptic genes from the prefrontal cortex of 12 primate species. The genes investigated included glutamate receptors, scaffolding proteins, synaptic vesicle components, as well as factors involved in synaptic vesicle release and structural components of the nervous system. Our analyses revealed that there have been significant changes during primate brain evolution in the components of the glutamatergic signaling pathway in terms of gene expression, protein expression, and promoter sequence changes. These results could entail functional modifications in the regulation of specific genes related to processes underlying learning and memory.
Collapse
Affiliation(s)
- Gerard Muntané
- Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| | - Julie E Horvath
- Department of Evolutionary Anthropology, Duke University, Durham, NC 27707, USA North Carolina Museum of Natural Sciences, Nature Research Center, Raleigh, NC 27601, USA Department of Biology, NC Central University, Durham, NC 27707, USA
| | - Patrick R Hof
- Fishberg Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 New York Consortium for Primate Evolution, New York, NY 10024, USA
| | - John J Ely
- Alamogordo Primate Facility, Holloman Air Force Base, NM 88330, USA
| | - William D Hopkins
- Neuroscience Institute and Language Research Center, Georgia State University, Atlanta, GA 30302, USA
| | - Mary Ann Raghanti
- Department of Anthropology and School of Biomedical Sciences, Kent State University, Kent, OH 44242, USA
| | | | - Gregory A Wray
- Department of Evolutionary Anthropology, Duke University, Durham, NC 27707, USA Biology Department, Duke University, Durham, NC, USA
| | - Chet C Sherwood
- Department of Anthropology, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
103
|
Marín-Padilla M. The mammalian neocortex new pyramidal neuron: a new conception. Front Neuroanat 2014; 7:51. [PMID: 24431992 PMCID: PMC3880895 DOI: 10.3389/fnana.2013.00051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 12/19/2013] [Indexed: 11/13/2022] Open
Abstract
The new cerebral cortex (neocortex) and the new type of pyramidal neuron are mammalian innovations that have evolved for operating their increasing motor capabilities while essentially using analogous anatomical and neural makeups. The human neocortex starts to develop in 6-week-old embryos with the establishment of a primordial cortical organization, which resembles the primitive cortices of amphibian and reptiles. From the 8th to the 15th week of age, new pyramidal neurons, of ependymal origin, are progressively incorporated within this primordial cortex forming a cellular plate that divides its components into those above it (neocortex first layer) and those below it (neocortex subplate zone). From the 16th week of age to birth and postnatally, the new pyramidal neurons continue to elongate functionally their apical dendrite by adding synaptic membrane to incorporate the needed sensory information for operating its developing motor activities. The new pyramidal neuron' distinguishing feature is the capacity of elongating anatomically and functionally its apical dendrite (its main receptive surface) without losing its original attachment to first layer or the location of its soma and, hence, retaining its essential nature. The number of pyramidal cell functional strata established in the motor cortex increases and reflects each mammalian species motor capabilities: the hedgehog needs two pyramidal cell functional strata to carry out all its motor activities, the mouse 3, cat 4, primates 5 and humans 6. The presence of six pyramidal cell functional strata distinguish the human motor cortex from that of others primates. Homo sapiens represent a new evolutionary stage that have transformed his primate brain for operating his unique motor capabilities, such as speaking, writing, painting, sculpturing and thinking as a premotor activity. Words used in language are the motor expression of thoughts and represent sounds produced by maneuvering the column of expiratory air by coordinated motor quivering as it passes through the larynx, pharynx, mouth, tongue, and lips. Homo sapiens cerebrum has developed new motor centers to communicate mental thoughts (and/or intention) through motor actions.
Collapse
Affiliation(s)
- Miguel Marín-Padilla
- Department of Pathology and Pediatrics, The Geisel School of Medicine at DartmouthHanover, NH USA
| |
Collapse
|
104
|
ArabidopsisGrowth Simulation Using Image Processing Technology. JOURNAL OF ELECTRICAL AND COMPUTER ENGINEERING 2014. [DOI: 10.1155/2014/751927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This paper aims to provide a method to represent the virtualArabidopsisplant at each growth stage. It includes simulating the shape and providing growth parameters. The shape is described with elliptic Fourier descriptors. First, the plant is segmented from the background with the chromatic coordinates. With the segmentation result, the outer boundary series are obtained by using boundary tracking algorithm. The elliptic Fourier analysis is then carried out to extract the coefficients of the contour. The coefficients require less storage than the original contour points and can be used to simulate the shape of the plant. The growth parameters include total area and the number of leaves of the plant. The total area is obtained with the number of the plant pixels and the image calibration result. The number of leaves is derived by detecting the apex of each leaf. It is achieved by using wavelet transform to identify the local maximum of the distance signal between the contour points and the region centroid. Experiment result shows that this method can record the growth stage ofArabidopsisplant with fewer data and provide a visual platform for plant growth research.
Collapse
|
105
|
Abstract
To understand the emergence of human higher cognition, we must understand its biological substrate--the cerebral cortex, which considers itself the crowning achievement of evolution. Here, we describe how advances in developmental neurobiology, coupled with those in genetics, including adaptive protein evolution via gene duplications and the emergence of novel regulatory elements, can provide insights into the evolutionary mechanisms culminating in the human cerebrum. Given that the massive expansion of the cortical surface and elaboration of its connections in humans originates from developmental events, understanding the genetic regulation of cell number, neuronal migration to proper layers, columns, and regions, and ultimately their differentiation into specific phenotypes, is critical. The pre- and postnatal environment also interacts with the cellular substrate to yield a basic network that is refined via selection and elimination of synaptic connections, a process that is prolonged in humans. This knowledge provides essential insight into the pathogenesis of human-specific neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | | |
Collapse
|
106
|
Shalhoub J, Sikkel MB, Davies KJ, Vorkas PA, Want EJ, Davies AH. Systems Biology of Human Atherosclerosis. Vasc Endovascular Surg 2013; 48:5-17. [DOI: 10.1177/1538574413510628] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Systems biology describes a holistic and integrative approach to understand physiology and pathology. The “omic” disciplines include genomics, transcriptomics, proteomics, and metabolic profiling (metabonomics and metabolomics). By adopting a stance, which is opposing (yet complimentary) to conventional research techniques, systems biology offers an overview by assessing the “net” biological effect imposed by a disease or nondisease state. There are a number of different organizational levels to be understood, from DNA to protein, metabolites, cells, organs and organisms, even beyond this to an organism’s context. Systems biology relies on the existence of “nodes” and “edges.” Nodes are the constituent part of the system being studied (eg, proteins in the proteome), while the edges are the way these constituents interact. In future, it will be increasingly important to collaborate, collating data from multiple studies to improve data sets, making them freely available and undertaking integrative analyses.
Collapse
Affiliation(s)
- Joseph Shalhoub
- Department of Surgery & Cancer, Academic Section of Vascular Surgery, Imperial College London, United Kingdom
| | - Markus B. Sikkel
- Myocardial Function Section, National Heart & Lung Institute, Imperial College London, United Kingdom
| | - Kerry J. Davies
- Department of Surgery & Cancer, Academic Section of Vascular Surgery, Imperial College London, United Kingdom
| | - Panagiotis A. Vorkas
- Department of Surgery & Cancer, Computational & Systems Medicine, Imperial College London, United Kingdom
| | - Elizabeth J. Want
- Department of Surgery & Cancer, Computational & Systems Medicine, Imperial College London, United Kingdom
| | - Alun H. Davies
- Department of Surgery & Cancer, Academic Section of Vascular Surgery, Imperial College London, United Kingdom
| |
Collapse
|
107
|
Differential L1 regulation in pluripotent stem cells of humans and apes. Nature 2013; 503:525-529. [PMID: 24153179 PMCID: PMC4064720 DOI: 10.1038/nature12686] [Citation(s) in RCA: 174] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 09/17/2013] [Indexed: 12/11/2022]
Abstract
Identifying cellular and molecular differences between human and non-human primates (NHPs) is essential to the basic understanding of the evolution and diversity of our own species. Until now, preserved tissues have been the main source for most comparative studies between humans, chimpanzees (Pan troglodytes) and bonobos (Pan paniscus)1,2. However, these tissue samples do not fairly represent the distinctive traits of live cell behavior and are not amenable to genetic manipulation. We hypothesized that induced pluripotent stem cells (iPSCs) could be a unique biological resource to elucidate relevant phenotypical differences between human and NHPs and that those differences could have potential adaptation and speciation value. Here, we describe the generation and initial characterization of iPSCs from chimpanzees and bonobos as novel tools to explore factors that have contributed to great ape evolution. Comparative gene expression analysis of human and NHP iPSCs revealed differences in the regulation of Long Interspersed Nuclear Element-1 (LINE-1 or L1) transposons. A force of change in mammalian evolution, L1 elements are retrotransposons that have remained active during primate evolution3-5. Decreased levels of L1 restricting factors APOBEC3B (A3B)6 and PIWIL27 in NHP iPSCs correlated with increased L1 mobility and endogenous L1 mRNA levels. Moreover, results from manipulation of A3B and PIWIL2 levels in iPSCs supported a causal inverse relationship between levels of these proteins and L1 retrotransposition. Finally, we found increased copy numbers of species-specific L1 elements in the genome of chimpanzees compared to humans, supporting the idea that increased L1 mobility in NHPs is not limited to iPSCs in culture and may have also occurred in the germline or embryonic cells developmentally upstream to germline specification during primate evolution. We propose that differences in L1 mobility may have differentially shaped the genomes of humans and NHPs and could have ongoing adaptive significance.
Collapse
|
108
|
Tong LJ, Flach EJ, Sheppard MN, Pocknell A, Banerjee AA, Boswood A, Bouts T, Routh A, Feltrer Y. Fatal arrhythmogenic right ventricular cardiomyopathy in 2 related subadult chimpanzees (Pan troglodytes). Vet Pathol 2013; 51:858-67. [PMID: 23988399 DOI: 10.1177/0300985813501333] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cardiovascular disease is increasingly recognized as an important cause of morbidity and mortality in captive chimpanzees (Pan troglodytes). This report records 2 cases of sudden cardiac death in closely related subadult captive chimpanzees with marked replacement fibrosis and adipocyte infiltration of the myocardium, which resemble specific atypical forms of the familial human disease arrhythmogenic right ventricular cardiomyopathy. Changes were consistent with left-dominant and biventricular subtypes, which are both phenotypic variants found within human families with familial arrhythmogenic right ventricular cardiomyopathy. Previously reported fibrosing cardiomyopathies in chimpanzees were characterized by nonspecific interstitial fibrosis, in contrast to the replacement fibrofatty infiltration with predilection for the outer myocardium seen in these 2 cases. To the authors' knowledge, this case report is the first to describe cardiomyopathy resembling arrhythmogenic right ventricular cardiomyopathy in nonhuman primates and the first to describe left-dominant arrhythmogenic cardiomyopathy-type lesions in an animal.
Collapse
Affiliation(s)
- L J Tong
- Zoological Society of London, Veterinary Department, London Zoo, Regents Park, London, UK Veterinary Pathology Diagnostic Services, Faculty of Veterinary Science, Sydney University, Australia
| | - E J Flach
- Zoological Society of London, Veterinary Department, London Zoo, Regents Park, London, UK
| | - M N Sheppard
- Cardiac Pathology Unit, Imperial College, London, UK
| | - A Pocknell
- Zoological Society of London, Veterinary Department, London Zoo, Regents Park, London, UK Finn Pathologists, One Eyed Lane, Weybread, Norfolk, UK
| | - A A Banerjee
- Royal Veterinary College, University of London, Hertfordshire, UK
| | - A Boswood
- Royal Veterinary College, University of London, Hertfordshire, UK
| | - T Bouts
- Whipsnade Zoo, Dunstable, Bedfordshire, UK Al Wabra Wildlife Preservation, Al Shahaniya, Qatar
| | - A Routh
- Zoological Society of London, Veterinary Department, London Zoo, Regents Park, London, UK Durrell Wildlife Conservation Trust, Jersey, Channel Islands
| | - Y Feltrer
- Zoological Society of London, Veterinary Department, London Zoo, Regents Park, London, UK
| |
Collapse
|
109
|
Duda P, Zrzavý J. Evolution of life history and behavior in Hominidae: towards phylogenetic reconstruction of the chimpanzee-human last common ancestor. J Hum Evol 2013; 65:424-46. [PMID: 23981863 DOI: 10.1016/j.jhevol.2013.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 07/29/2013] [Accepted: 07/29/2013] [Indexed: 02/08/2023]
Abstract
The origin of the fundamental behavioral differences between humans and our closest living relatives is one of the central issues of evolutionary anthropology. The prominent, chimpanzee-based referential model of early hominin behavior has recently been challenged on the basis of broad multispecies comparisons and newly discovered fossil evidence. Here, we argue that while behavioral data on extant great apes are extremely relevant for reconstruction of ancestral behaviors, these behaviors should be reconstructed trait by trait using formal phylogenetic methods. Using the widely accepted hominoid phylogenetic tree, we perform a series of character optimization analyses using 65 selected life-history and behavioral characters for all extant hominid species. This analysis allows us to reconstruct the character states of the last common ancestors of Hominoidea, Hominidae, and the chimpanzee-human last common ancestor. Our analyses demonstrate that many fundamental behavioral and life-history attributes of hominids (including humans) are evidently ancient and likely inherited from the common ancestor of all hominids. However, numerous behaviors present in extant great apes represent their own terminal autapomorphies (both uniquely derived and homoplastic). Any evolutionary model that uses a single extant species to explain behavioral evolution of early hominins is therefore of limited use. In contrast, phylogenetic reconstruction of ancestral states is able to provide a detailed suite of behavioral, ecological and life-history characters for each hypothetical ancestor. The living great apes therefore play an important role for the confident identification of the traits found in the chimpanzee-human last common ancestor, some of which are likely to represent behaviors of the fossil hominins.
Collapse
Affiliation(s)
- Pavel Duda
- Department of Zoology, Faculty of Science, University of South Bohemia, Branišovská 31, 370 05 České Budĕjovice, Czech Republic.
| | | |
Collapse
|
110
|
Paixão-Côrtes VR, Viscardi LH, Salzano FM, Cátira Bortolini M, Hünemeier T. The cognitive ability of extinct hominins: bringing down the hierarchy using genomic evidences. Am J Hum Biol 2013; 25:702-5. [PMID: 23907779 DOI: 10.1002/ajhb.22426] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 06/03/2013] [Accepted: 06/11/2013] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The availability of the full genomes of Homo sapiens, Homo neanderthalensis, and Denisovans, as well as modern bioinformatic tools, are opening new possibilities for the understanding of the differences and similarities present in these taxa. METHODS We searched for cognitive genes, examined their status in the genomes of these three entities. All substitutions present among them were retrieved. RESULTS We found 93 nonsynonymous substitutions in 51 cognitive genes, in which the derived allele was present in archaic and modern humans and the ancestral allele in other nonhuman primates. CONCLUSIONS The general picture obtained is of similarity in cognitive genes between extinct and extant humans.
Collapse
Affiliation(s)
- Vanessa R Paixão-Côrtes
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | | | |
Collapse
|
111
|
Quach H, Wilson D, Laval G, Patin E, Manry J, Guibert J, Barreiro LB, Nerrienet E, Verschoor E, Gessain A, Przeworski M, Quintana-Murci L. Different selective pressures shape the evolution of Toll-like receptors in human and African great ape populations. Hum Mol Genet 2013; 22:4829-40. [PMID: 23851028 DOI: 10.1093/hmg/ddt335] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The study of the genetic and selective landscape of immunity genes across primates can provide insight into the existing differences in susceptibility to infection observed between human and non-human primates. Here, we explored how selection has driven the evolution of a key family of innate immunity receptors, the Toll-like receptors (TLRs), in African great ape species. We sequenced the 10 TLRs in various populations of chimpanzees and gorillas, and analysed these data jointly with a human data set. We found that purifying selection has been more pervasive in great apes than in humans. Furthermore, in chimpanzees and gorillas, purifying selection has targeted TLRs irrespectively of whether they are endosomal or cell surface, in contrast to humans where strong selective constraints are restricted to endosomal TLRs. These observations suggest important differences in the relative importance of TLR-mediated pathogen sensing, such as that of recognition of flagellated bacteria by TLR5, between humans and great apes. Lastly, we used a population genetics-phylogenetics method that jointly analyses polymorphism and divergence data to detect fine-scale variation in selection pressures at specific codons within TLR genes. We identified different codons at different TLRs as being under positive selection in each species, highlighting that functional variation at these genes has conferred a selective advantage in immunity to infection to specific primate species. Overall, this study showed that the degree of selection driving the evolution of TLRs has largely differed between human and non-human primates, increasing our knowledge on their respective biological contribution to host defence in the natural setting.
Collapse
|
112
|
Paixão-Côrtes VR, Viscardi LH, Salzano FM, Hünemeier T, Bortolini MC. Homo sapiens, Homo neanderthalensis and the Denisova specimen: New insights on their evolutionary histories using whole-genome comparisons. Genet Mol Biol 2012; 35:904-11. [PMID: 23413113 PMCID: PMC3571422 DOI: 10.1590/s1415-47572012000600003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
After a brief review of the most recent findings in the study of human evolution, an extensive comparison of the complete genomes of our nearest relative, the chimpanzee (Pan troglodytes), of extant Homo sapiens, archaic Homo neanderthalensis and the Denisova specimen were made. The focus was on non-synonymous mutations, which consequently had an impact on protein levels and these changes were classified according to degree of effect. A total of 10,447 non-synonymous substitutions were found in which the derived allele is fixed or nearly fixed in humans as compared to chimpanzee. Their most frequent location was on chromosome 21. Their presence was then searched in the two archaic genomes. Mutations in 381 genes would imply radical amino acid changes, with a fraction of these related to olfaction and other important physiological processes. Eight new alleles were identified in the Neanderthal and/or Denisova genetic pools. Four others, possibly affecting cognition, occured both in the sapiens and two other archaic genomes. The selective sweep that gave rise to Homo sapiens could, therefore, have initiated before the modern/archaic human divergence.
Collapse
Affiliation(s)
| | - Lucas Henrique Viscardi
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Francisco Mauro Salzano
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Tábita Hünemeier
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Maria Cátira Bortolini
- Departamento de Genética, Instituto de Biociências, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| |
Collapse
|
113
|
Nagy Z, Szabó DR, Zsippai A, Falus A, Rácz K, Igaz P. [Relevance of long non-coding RNAs in tumour biology]. Orv Hetil 2012; 153:1494-501. [PMID: 22985664 DOI: 10.1556/oh.2012.29462] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The discovery of the biological relevance of non-coding RNA molecules represents one of the most significant advances in contemporary molecular biology. It has turned out that a major fraction of the non-coding part of the genome is transcribed. Beside small RNAs (including microRNAs) more and more data are disclosed concerning long non-coding RNAs of 200 nucleotides to 100 kb length that are implicated in the regulation of several basic molecular processes (cell proliferation, chromatin functioning, microRNA-mediated effects, etc.). Some of these long non-coding RNAs have been associated with human tumours, including H19, HOTAIR, MALAT1, etc., the different expression of which has been noted in various neoplasms relative to healthy tissues. Long non-coding RNAs may represent novel markers of molecular diagnostics and they might even turn out to be targets of therapeutic intervention.
Collapse
Affiliation(s)
- Zoltán Nagy
- Semmelweis Egyetem, Általános Orvostudományi Kar II. Belgyógyászati Klinika Budapest
| | | | | | | | | | | |
Collapse
|
114
|
Kim DS, Hahn Y. Gains of ubiquitylation sites in highly conserved proteins in the human lineage. BMC Bioinformatics 2012; 13:306. [PMID: 23157318 PMCID: PMC3561281 DOI: 10.1186/1471-2105-13-306] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2012] [Accepted: 11/14/2012] [Indexed: 01/03/2023] Open
Abstract
Background Post-translational modification of lysine residues of specific proteins by ubiquitin modulates the degradation, localization, and activity of these target proteins. Here, we identified gains of ubiquitylation sites in highly conserved regions of human proteins that occurred during human evolution. Results We analyzed human ubiquitylation site data and multiple alignments of orthologous mammalian proteins including those from humans, primates, other placental mammals, opossum, and platypus. In our analysis, we identified 281 ubiquitylation sites in 252 proteins that first appeared along the human lineage during primate evolution: one protein had four novel sites; four proteins had three sites each; 18 proteins had two sites each; and the remaining 229 proteins had one site each. PML, which is involved in neurodevelopment and neurodegeneration, acquired three sites, two of which have been reported to be involved in the degradation of PML. Thirteen human proteins, including ERCC2 (also known as XPD) and NBR1, gained human-specific ubiquitylated lysines after the human-chimpanzee divergence. ERCC2 has a Lys/Gln polymorphism, the derived (major) allele of which confers enhanced DNA repair capacity and reduced cancer risk compared with the ancestral (minor) allele. NBR1 and eight other proteins that are involved in the human autophagy protein interaction network gained a novel ubiquitylation site. Conclusions The gain of novel ubiquitylation sites could be involved in the evolution of protein degradation and other regulatory networks. Although gains of ubiquitylation sites do not necessarily equate to adaptive evolution, they are useful candidates for molecular functional analyses to identify novel advantageous genetic modifications and innovative phenotypes acquired during human evolution.
Collapse
Affiliation(s)
- Dong Seon Kim
- Department of Life Science, Research Center for Biomolecules and Biosystems, Chung-Ang University, Seoul, 156-756, Korea.
| | | |
Collapse
|
115
|
Kim DS, Hahn Y. Human-specific protein isoforms produced by novel splice sites in the human genome after the human-chimpanzee divergence. BMC Bioinformatics 2012; 13:299. [PMID: 23148531 PMCID: PMC3538075 DOI: 10.1186/1471-2105-13-299] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2012] [Accepted: 11/09/2012] [Indexed: 11/16/2022] Open
Abstract
Background Evolution of splice sites is a well-known phenomenon that results in transcript diversity during human evolution. Many novel splice sites are derived from repetitive elements and may not contribute to protein products. Here, we analyzed annotated human protein-coding exons and identified human-specific splice sites that arose after the human-chimpanzee divergence. Results We analyzed multiple alignments of the annotated human protein-coding exons and their respective orthologous mammalian genome sequences to identify 85 novel splice sites (50 splice acceptors and 35 donors) in the human genome. The novel protein-coding exons, which are expressed either constitutively or alternatively, produce novel protein isoforms by insertion, deletion, or frameshift. We found three cases in which the human-specific isoform conferred novel molecular function in the human cells: the human-specific IMUP protein isoform induces apoptosis of the trophoblast and is implicated in pre-eclampsia; the intronization of a part of SMOX gene exon produces inactive spermine oxidase; the human-specific NUB1 isoform shows reduced interaction with ubiquitin-like proteins, possibly affecting ubiquitin pathways. Conclusions Although the generation of novel protein isoforms does not equate to adaptive evolution, we propose that these cases are useful candidates for a molecular functional study to identify proteomic changes that might bring about novel phenotypes during human evolution.
Collapse
Affiliation(s)
- Dong Seon Kim
- Department of Life Science, Research Center for Biomolecules and Biosystems, Chung-Ang University, Seoul 156-756, Korea
| | | |
Collapse
|
116
|
A "forward genomics" approach links genotype to phenotype using independent phenotypic losses among related species. Cell Rep 2012; 2:817-23. [PMID: 23022484 DOI: 10.1016/j.celrep.2012.08.032] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 07/31/2012] [Accepted: 08/30/2012] [Indexed: 12/27/2022] Open
Abstract
Genotype-phenotype mapping is hampered by countless genomic changes between species. We introduce a computational "forward genomics" strategy that-given only an independently lost phenotype and whole genomes-matches genomic and phenotypic loss patterns to associate specific genomic regions with this phenotype. We conducted genome-wide screens for two metabolic phenotypes. First, our approach correctly matches the inactivated Gulo gene exactly with the species that lost the ability to synthesize vitamin C. Second, we attribute naturally low biliary phospholipid levels in guinea pigs and horses to the inactivated phospholipid transporter Abcb4. Human ABCB4 mutations also result in low phospholipid levels but lead to severe liver disease, suggesting compensatory mechanisms in guinea pig and horse. Our simulation studies, counts of independent changes in existing phenotype surveys, and the forthcoming availability of many new genomes all suggest that forward genomics can be applied to many phenotypes, including those relevant for human evolution and disease.
Collapse
|
117
|
Abstract
BACKGROUND The concept of conserved processes presents unique opportunities for using nonhuman animal models in biomedical research. However, the concept must be examined in the context that humans and nonhuman animals are evolved, complex, adaptive systems. Given that nonhuman animals are examples of living systems that are differently complex from humans, what does the existence of a conserved gene or process imply for inter-species extrapolation? METHODS We surveyed the literature including philosophy of science, biological complexity, conserved processes, evolutionary biology, comparative medicine, anti-neoplastic agents, inhalational anesthetics, and drug development journals in order to determine the value of nonhuman animal models when studying conserved processes. RESULTS Evolution through natural selection has employed components and processes both to produce the same outcomes among species but also to generate different functions and traits. Many genes and processes are conserved, but new combinations of these processes or different regulation of the genes involved in these processes have resulted in unique organisms. Further, there is a hierarchy of organization in complex living systems. At some levels, the components are simple systems that can be analyzed by mathematics or the physical sciences, while at other levels the system cannot be fully analyzed by reducing it to a physical system. The study of complex living systems must alternate between focusing on the parts and examining the intact whole organism while taking into account the connections between the two. Systems biology aims for this holism. We examined the actions of inhalational anesthetic agents and anti-neoplastic agents in order to address what the characteristics of complex living systems imply for inter-species extrapolation of traits and responses related to conserved processes. CONCLUSION We conclude that even the presence of conserved processes is insufficient for inter-species extrapolation when the trait or response being studied is located at higher levels of organization, is in a different module, or is influenced by other modules. However, when the examination of the conserved process occurs at the same level of organization or in the same module, and hence is subject to study solely by reductionism, then extrapolation is possible.
Collapse
Affiliation(s)
- Ray Greek
- Americans For Medical Advancement (www.AFMA-curedisease.org), 2251 Refugio Rd, Goleta, CA, 93117, USA
| | - Mark J Rice
- Department of Anesthesiology, University of Florida College of Medicine, PO Box 100254, Gainesville, FL, 32610-0254, USA
| |
Collapse
|
118
|
Wang J, Cao X, Zhang Y, Su B. Genome-wide DNA methylation analyses in the brain reveal four differentially methylated regions between humans and non-human primates. BMC Evol Biol 2012; 12:144. [PMID: 22899776 PMCID: PMC3483258 DOI: 10.1186/1471-2148-12-144] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 08/03/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The highly improved cognitive function is the most significant change in human evolutionary history. Recently, several large-scale studies reported the evolutionary roles of DNA methylation; however, the role of DNA methylation on brain evolution is largely unknown. RESULTS To test if DNA methylation has contributed to the evolution of human brain, with the use of MeDIP-Chip and SEQUENOM MassARRAY, we conducted a genome-wide analysis to identify differentially methylated regions (DMRs) in the brain between humans and rhesus macaques. We first identified a total of 150 candidate DMRs by the MeDIP-Chip method, among which 4 DMRs were confirmed by the MassARRAY analysis. All 4 DMRs are within or close to the CpG islands, and a MIR3 repeat element was identified in one DMR, but no repeat sequence was observed in the other 3 DMRs. For the 4 DMR genes, their proteins tend to be conserved and two genes have neural related functions. Bisulfite sequencing and phylogenetic comparison among human, chimpanzee, rhesus macaque and rat suggested several regions of lineage specific DNA methylation, including a human specific hypomethylated region in the promoter of K6IRS2 gene. CONCLUSIONS Our study provides a new angle of studying human brain evolution and understanding the evolutionary role of DNA methylation in the central nervous system. The results suggest that the patterns of DNA methylation in the brain are in general similar between humans and non-human primates, and only a few DMRs were identified.
Collapse
Affiliation(s)
- Jinkai Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, People's Republic of China
| | | | | | | |
Collapse
|
119
|
Wu DD, Li GM, Jin W, Li Y, Zhang YP. Positive selection on the osteoarthritis-risk and decreased-height associated variants at the GDF5 gene in East Asians. PLoS One 2012; 7:e42553. [PMID: 22905146 PMCID: PMC3419199 DOI: 10.1371/journal.pone.0042553] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/10/2012] [Indexed: 11/19/2022] Open
Abstract
GDF5 is a member of the bone morphogenetic protein (BMP) gene family, and plays an important role in the development of the skeletal system. Variants of the gene are associated with osteoarthritis and height in some human populations. Here, we resequenced the gene in individuals from four geographically separated human populations, and found that the evolution of the promoter region deviated from neutral expectations, with the sequence evolution driven by positive selection in the East Asian population, especially the haplotypes carrying the derived alleles of 5' UTR SNPs rs143384 and rs143383. The derived alleles of rs143384 and rs143383, which are associated with a risk of osteoarthritis and decreased height, have high frequencies in non-Africans and show strong extended haplotype homozygosity and high population differentiation in East Asian. It is concluded that positive selection has driven the rapid evolution of the two osteoarthritis osteoarthritis-risk and decreased height associated variants of the human GDF5 gene, and supports the suggestion that the reduction in body size during the terminal Pleistocene and Holocene period might have been an adaptive process influenced by genetic factors.
Collapse
Affiliation(s)
- Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Gui-Mei Li
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| | - Wei Jin
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| | - Yan Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| |
Collapse
|
120
|
Schneider E, Mayer S, El Hajj N, Jensen LR, Kuss AW, Zischler H, Kondova I, Bontrop RE, Navarro B, Fuchs E, Zechner U, Haaf T. Methylation and expression analyses of the 7q autism susceptibility locus genes MEST , COPG2, and TSGA14 in human and anthropoid primate cortices. Cytogenet Genome Res 2012; 136:278-87. [PMID: 22456293 DOI: 10.1159/000337298] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/30/2012] [Indexed: 12/11/2022] Open
Abstract
The autism susceptibility locus on human chromosome 7q32 contains the maternally imprinted MEST and the non-imprinted COPG2 and TSGA14 genes. Autism is a disorder of the 'social brain' that has been proposed to be due to an overbalance of paternally expressed genes. To study regulation of the 7q32 locus during anthropoid primate evolution, we analyzed the methylation and expression patterns of MEST, COPG2, and TSGA14 in human, chimpanzee, Old World monkey (baboon and rhesus macaque), and New World monkey (marmoset) cortices. In all human and anthropoid primate cortices, the MEST promoter was hemimethylated, as expected for a differentially methylated imprinting control region, whereas the COPG2 and TSGA14 promoters were completely demethylated, typical for transcriptionally active non-imprinted genes. The MEST gene also showed comparable mRNA expression levels in all analyzed species. In contrast, COPG2 expression was downregulated in the human cortex compared to chimpanzee, Old and New World monkeys. TSGA14 either showed no differential regulation in the human brain compared to chimpanzee and marmoset or a slight upregulation compared to baboon. The human-specific downregulation supports a role for COPG2 in the development of a 'social brain'. Promoter methylation patterns appear to be more stable during evolution than gene expression patterns, suggesting that other mechanisms may be more important for inter-primate differences in gene expression.
Collapse
Affiliation(s)
- E Schneider
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Bailey J. Lessons from chimpanzee-based research on human disease: the implications of genetic differences. Altern Lab Anim 2012; 39:527-40. [PMID: 22243397 DOI: 10.1177/026119291103900608] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Assertions that the use of chimpanzees to investigate human diseases is valid scientifically are frequently based on a reported 98-99% genetic similarity between the species. Critical analyses of the relevance of chimpanzee studies to human biology, however, indicate that this genetic similarity does not result in sufficient physiological similarity for the chimpanzee to constitute a good model for research, and furthermore, that chimpanzee data do not translate well to progress in clinical practice for humans. Leading examples include the minimal citations of chimpanzee research that is relevant to human medicine, the highly different pathology of HIV/AIDS and hepatitis C virus infection in the two species, the lack of correlation in the efficacy of vaccines and treatments between chimpanzees and humans, and the fact that chimpanzees are not useful for research on human cancer. The major molecular differences underlying these inter-species phenotypic disparities have been revealed by comparative genomics and molecular biology - there are key differences in all aspects of gene expression and protein function, from chromosome and chromatin structure to post-translational modification. The collective effects of these differences are striking, extensive and widespread, and they show that the superficial similarity between human and chimpanzee genetic sequences is of little consequence for biomedical research. The extrapolation of biomedical data from the chimpanzee to the human is therefore highly unreliable, and the use of the chimpanzee must be considered of little value, particularly given the breadth and potential of alternative methods of enquiry that are currently available to science.
Collapse
|
122
|
Schneider E, Jensen LR, Farcas R, Kondova I, Bontrop RE, Navarro B, Fuchs E, Kuss AW, Haaf T. A high density of human communication-associated genes in chromosome 7q31-q36: differential expression in human and non-human primate cortices. Cytogenet Genome Res 2012; 136:97-106. [PMID: 22261840 DOI: 10.1159/000335465] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2011] [Indexed: 11/19/2022] Open
Abstract
The human brain is distinguished by its remarkable size, high energy consumption, and cognitive abilities compared to all other mammals and non-human primates. However, little is known about what has accelerated brain evolution in the human lineage. One possible explanation is that the appearance of advanced communication skills and language has been a driving force of human brain development. The phenotypic adaptations in brain structure and function which occurred on the way to modern humans may be associated with specific molecular signatures in today's human genome and/or transcriptome. Genes that have been linked to language, reading, and/or autism spectrum disorders are prime candidates when searching for genes for human-specific communication abilities. The database and genome-wide expression analyses we present here revealed a clustering of such communication-associated genes (COAG) on human chromosomes X and 7, in particular chromosome 7q31-q36. Compared to the rest of the genome, we found a high number of COAG to be differentially expressed in the cortices of humans and non-human primates (chimpanzee, baboon, and/or marmoset). The role of X-linked genes for the development of human-specific cognitive abilities is well known. We now propose that chromosome 7q31-q36 also represents a hot spot for the evolution of human-specific communication abilities. Selective pressure on the T cell receptor beta locus on chromosome 7q34, which plays a pivotal role in the immune system, could have led to rapid dissemination of positive gene variants in hitchhiking COAG.
Collapse
Affiliation(s)
- E Schneider
- Institute of Human Genetics, Julius Maximilians University, Würzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
123
|
Human-Specific Changes in Sialic Acid Biology. POST-GENOME BIOLOGY OF PRIMATES 2012. [PMCID: PMC7120309 DOI: 10.1007/978-4-431-54011-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Sialic acids are components of cell-surface glycans and play important roles in cell–cell communication and host–pathogen interaction. More than 55 genes, encoding receptors, enzymes, and transporters, are known to be involved in sialic acid biology. Nearly 10 years of research have revealed that several of these genes show human-specific changes in genome structure, expression, or function. In this chapter, we introduce these human-specific changes and their possible impact on the human evolution. Also, we give an overview of the evolution of sialic acid biology in primates. The discovery of human-specific changes in sialic acid biology is one step toward explaining the genetic basis of human uniqueness, one of the major activities in primatology, contributing to answering a transdisciplinary question: What makes us human?
Collapse
|
124
|
Affiliation(s)
- Daniele Guerzoni
- Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, Ireland
| | - Aoife McLysaght
- Smurfit Institute of Genetics, University of Dublin, Trinity College, Dublin, Ireland
- * E-mail:
| |
Collapse
|
125
|
Polavarapu N, Arora G, Mittal VK, McDonald JF. Characterization and potential functional significance of human-chimpanzee large INDEL variation. Mob DNA 2011; 2:13. [PMID: 22024410 PMCID: PMC3215961 DOI: 10.1186/1759-8753-2-13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/25/2011] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Although humans and chimpanzees have accumulated significant differences in a number of phenotypic traits since diverging from a common ancestor about six million years ago, their genomes are more than 98.5% identical at protein-coding loci. This modest degree of nucleotide divergence is not sufficient to explain the extensive phenotypic differences between the two species. It has been hypothesized that the genetic basis of the phenotypic differences lies at the level of gene regulation and is associated with the extensive insertion and deletion (INDEL) variation between the two species. To test the hypothesis that large INDELs (80 to 12,000 bp) may have contributed significantly to differences in gene regulation between the two species, we categorized human-chimpanzee INDEL variation mapping in or around genes and determined whether this variation is significantly correlated with previously determined differences in gene expression. RESULTS Extensive, large INDEL variation exists between the human and chimpanzee genomes. This variation is primarily attributable to retrotransposon insertions within the human lineage. There is a significant correlation between differences in gene expression and large human-chimpanzee INDEL variation mapping in genes or in proximity to them. CONCLUSIONS The results presented herein are consistent with the hypothesis that large INDELs, particularly those associated with retrotransposons, have played a significant role in human-chimpanzee regulatory evolution.
Collapse
Affiliation(s)
| | - Gaurav Arora
- Parker H Petit Institute for Bioengineering & Bioscience, School of Biology, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - Vinay K Mittal
- Parker H Petit Institute for Bioengineering & Bioscience, School of Biology, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| | - John F McDonald
- Parker H Petit Institute for Bioengineering & Bioscience, School of Biology, Georgia Institute of Technology, 315 Ferst Drive NW, Atlanta, GA 30332, USA
| |
Collapse
|
126
|
Suzuki K, Tanigawa K, Kawashima A, Miyamura T, Ishii N. Chimpanzees used for medical research shed light on the pathoetiology of leprosy. Future Microbiol 2011; 6:1151-7. [PMID: 22004034 DOI: 10.2217/fmb.11.97] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Leprosy is a chronic infectious disorder caused by Mycobacterium leprae, which mainly affects skin and peripheral nerves. It is classified as either paucibacillary or multibacillary based upon clinical manifestations and slit-skin smear results. It is speculated that leprosy develops after a long latency period following M. leprae infection. However, the actual time of infection and the duration of latency have never been proven in human patients. To date, four cases of spontaneous leprosy have been reported in chimpanzees who were caught in West Africa in infancy and used for medical research in the USA and Japan. One of these chimpanzees was extensively studied in Japan, and single-nucleotide polymorphism analysis for the M. leprae genome was conducted. This analysis revealed that the chimpanzee was infected with M. leprae during infancy in West Africa and the pathognomonic signs of leprosy appeared after at least 30 years of incubation. Analysis of leprosy in chimpanzees can contribute not only to medical research but also to the understanding of the pathoetiology of leprosy.
Collapse
Affiliation(s)
- Koichi Suzuki
- Laboratory of Molecular Diagnostics, Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, 4-2-1 Aoba-cho, Higashimurayama, Tokyo 189-0002, Japan.
| | | | | | | | | |
Collapse
|
127
|
Abstract
The study of human and primate altruism faces an evolutionary anomaly: There is ample evidence for altruistic preferences in our own species and growing evidence in monkeys, but one of our closest relatives, the chimpanzee (Pan troglodytes), is viewed as a reluctant altruist, acting only in response to pressure and solicitation. Although chimpanzee prosocial behavior has been reported both in observational captive studies and in the wild, thus far Prosocial Choice Tests have failed to produce evidence. However, methodologies of previous Prosocial Choice Tests may have handicapped the apes unintentionally. Here we present findings of a paradigm in which chimpanzees chose between two differently colored tokens: one "selfish" token resulting in a reward for the actor only (1/0), and the other "prosocial" token rewarding both the actor and a partner (1/1). Seven female chimpanzees, each tested with three different partners, showed a significant bias for the prosocial option. Prosocial choices occurred both in response to solicitation by the partner and spontaneously without solicitation. However, directed requests and pressure by the partner reduced the actor's prosocial tendency. These results draw into question previous conclusions indicating that chimpanzees have a limited sensitivity to the needs of others and behave prosocially only in response to significant prompting.
Collapse
|
128
|
Kim DS, Hahn Y. Identification of novel phosphorylation modification sites in human proteins that originated after the human-chimpanzee divergence. ACTA ACUST UNITED AC 2011; 27:2494-501. [PMID: 21775310 DOI: 10.1093/bioinformatics/btr426] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
MOTIVATION Phosphorylation modifications of specific protein residues are involved in a wide range of biological processes such as modulation of intracellular signal networks. Here, we present the development and application of a bioinformatics procedure for systematic identification of human-specific phosphorylation sites in proteins that may have occurred after the human-chimpanzee divergence. RESULTS We collected annotated human phosphorylation sites and compared each site to orthologous mammalian proteins across taxa including chimpanzee, orangutan, rhesus macaque, marmoset, mouse, dog, cow, elephant, opossum and platypus. We identified 37 human-specific gains of annotated phosphorylation sites in 35 proteins: 22 serines, 12 threonines and 3 tyrosines. The novel phosphorylation sites are situated in highly conserved segments of the protein. Proteins with novel phosphorylation sites are involved in crucial biological processes such as cell division (AURKB, CASC5, MKI67 and PDCD4) and chromatin remodeling (HIRA, HIRIP3, HIST1H1T, NAP1L4 and LRWD1). Modified phosphorylatable residues produce novel target sites for protein kinases such as cyclin-dependent kinases and casein kinases, possibly resulting in rewiring and fine-tuning of phosphorylation regulatory networks. The potential human-specific phosphorylation sites identified in this study are useful as candidates for functional analysis to identify novel phenotypes in humans. CONTACT hahny@cau.ac.kr SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Dong Seon Kim
- School of Biological Sciences (BK21 Program) and Research Center for Biomolecules and Biosystems, Chung-Ang University, Seoul 156-756, Korea
| | | |
Collapse
|
129
|
Bakacs T, Mehrishi JN, Moss RW. Ipilimumab (Yervoy) and the TGN1412 catastrophe. Immunobiology 2011; 217:583-9. [PMID: 21821307 DOI: 10.1016/j.imbio.2011.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Accepted: 07/03/2011] [Indexed: 12/12/2022]
Abstract
The development of the anti-CTLA-4 antibody (ipilimumab; marketed as Yervoy) immune regulatory therapy was based on the premise that "Abrogation of the function of CTLA-4 would permit CD28 to function unopposed and might swing the balance in favor of immune stimulation, tolerance breakdown and tumor eradication…" (Weber, 2009). By now, the vast majority of data collected from more than 4000 patients proves that this prediction was entirely correct. Paradoxically, the successful blockade of immune checkpoints raises the question whether an anti-CTLA-4 antibody could ever become an important therapy against cancer. T cells lost their ability to discriminate between self and non-self. Thus, tolerance to self tissues was broken in ∼70% of the patients. In the recent industry-sponsored phase III clinical trial of ipilimumab, 147 (38.7%) of the patients experienced severe adverse events and 6.8% suffered dose-limiting events (8.4%, in the ipilimumab-alone group). There were 14 deaths related to the study drugs and 7 of these were associated with immune-related adverse events. In contrast, the complete response rate was only 0.2%, in one patient out of 403 who received ipilimumab plus a peptide vaccine. Promoters of ipilimumab appear to be unmindful of the clinical trial catastrophe in London. Then, a humanized "superagonist" anti-CD28 monoclonal antibody, TGN1412, which "preferentially" activated regulatory T cells, at a higher dose, also activated all CD28 positive T cells. This precipitated a "cytokine storm" leading to life-threatening multiple organ failure in the six healthy human volunteers. Neither anti-CD28 nor anti-CTLA-4 therapies rely on antigen-specificity. Both release free antibody into the body against common molecular targets that are expressed on the targeted as well as on the non-targeted T cells. At lower antibody doses specific T cells are preferentially activated. With increasing antibody dose, however, the kinetics of the interaction is pushed in favor of widespread non-specific T cell expansion. Using the law of mass action we calculated that the vast majority of the CTLA-4 receptors on all activated T cells (including melanoma specific T cells) in the phase III clinical trial of ipilimumab will have been saturated. This would explain the runaway immune response observed. The conclusions drawn by the authors of the ipilimumab trial paper could bear an independent inspection and reassessment concerning the validation of the blockade of immune checkpoints as an important therapeutic strategy against cancer.
Collapse
Affiliation(s)
- Tibor Bakacs
- Department of Probability, Alfred Renyi Institute of Mathematics, Hungarian Academy of Sciences, Realtanoda utca 13-15, H-1053 Budapest, Hungary.
| | | | | |
Collapse
|
130
|
Mitra N, Banda K, Altheide TK, Schaffer L, Johnson-Pais TL, Beuten J, Leach RJ, Angata T, Varki N, Varki A. SIGLEC12, a human-specific segregating (pseudo)gene, encodes a signaling molecule expressed in prostate carcinomas. J Biol Chem 2011; 286:23003-11. [PMID: 21555517 PMCID: PMC3123068 DOI: 10.1074/jbc.m111.244152] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 05/05/2011] [Indexed: 12/15/2022] Open
Abstract
The primate SIGLEC12 gene encodes one of the CD33-related Siglec family of signaling molecules in immune cells. We had previously reported that this gene harbors a human-specific missense mutation of the codon for an Arg residue required for sialic acid recognition. Here we show that this R122C mutation of the Siglec-XII protein is fixed in the human population, i.e. it occurred prior to the origin of modern humans. Additional mutations have since completely inactivated the SIGLEC12 gene in some but not all humans. The most common inactivating mutation with a global allele frequency of 58% is a single nucleotide frameshift that markedly shortens the open reading frame. Unlike other CD33-related Siglecs that are primarily found on immune cells, we found that Siglec-XII protein is expressed not only on some macrophages but also on various epithelial cell surfaces in humans and chimpanzees. We also found expression on certain human prostate epithelial carcinomas and carcinoma cell lines. This expression correlates with the presence of the nonframeshifted, intact SIGLEC12 allele. Although SIGLEC12 allele status did not predict prostate carcinoma incidence, restoration of expression in a prostate carcinoma cell line homozygous for the frameshift mutation induced altered regulation of several genes associated with carcinoma progression. These stably transfected Siglec-XII-expressing prostate cancer cells also showed enhanced growth in nude mice. Finally, monoclonal antibodies against the protein were internalized by Siglec-XII-expressing prostate carcinoma cells, allowing targeting of a toxin to such cells. Polymorphic expression of Siglec-XII in humans thus has implications for prostate cancer biology and therapeutics.
Collapse
Affiliation(s)
- Nivedita Mitra
- From the Glycobiology Research and Training Center, Departments of Medicine, Pathology and Cellular & Molecular Medicine, University of California at San Diego, La Jolla, California 92093
| | - Kalyan Banda
- From the Glycobiology Research and Training Center, Departments of Medicine, Pathology and Cellular & Molecular Medicine, University of California at San Diego, La Jolla, California 92093
| | - Tasha K. Altheide
- From the Glycobiology Research and Training Center, Departments of Medicine, Pathology and Cellular & Molecular Medicine, University of California at San Diego, La Jolla, California 92093
| | - Lana Schaffer
- the Scripps Research Institute, La Jolla, California 92037, and
| | - Teresa L. Johnson-Pais
- the Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Joke Beuten
- the Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Robin J. Leach
- the Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Takashi Angata
- From the Glycobiology Research and Training Center, Departments of Medicine, Pathology and Cellular & Molecular Medicine, University of California at San Diego, La Jolla, California 92093
| | - Nissi Varki
- From the Glycobiology Research and Training Center, Departments of Medicine, Pathology and Cellular & Molecular Medicine, University of California at San Diego, La Jolla, California 92093
| | - Ajit Varki
- From the Glycobiology Research and Training Center, Departments of Medicine, Pathology and Cellular & Molecular Medicine, University of California at San Diego, La Jolla, California 92093
| |
Collapse
|
131
|
Abstract
The past two decades have witnessed tremendous advances in noninvasive and postmortem neuroscientific techniques, advances that have made it possible, for the first time, to compare in detail the organization of the human brain to that of other primates. Studies comparing humans to chimpanzees and other great apes reveal that human brain evolution was not merely a matter of enlargement, but involved changes at all levels of organization that have been examined. These include the cellular and laminar organization of cortical areas; the higher order organization of the cortex, as reflected in the expansion of association cortex (in absolute terms, as well as relative to primary areas); the distribution of long-distance cortical connections; and hemispheric asymmetry. Additionally, genetic differences between humans and other primates have proven to be more extensive than previously thought, raising the possibility that human brain evolution involved significant modifications of neurophysiology and cerebral energy metabolism.
Collapse
Affiliation(s)
- Todd M Preuss
- Division of Neuropathology and Neurodegenerative Diseases and Center for Translational Social Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329, USA.
| |
Collapse
|
132
|
Wang X, Chow R, Deng L, Anderson D, Weidner N, Godwin AK, Bewtra C, Zlotnik A, Bui J, Varki A, Varki N. Expression of Siglec-11 by human and chimpanzee ovarian stromal cells, with uniquely human ligands: implications for human ovarian physiology and pathology. Glycobiology 2011; 21:1038-48. [PMID: 21467073 DOI: 10.1093/glycob/cwr039] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Siglecs (Sialic acid-binding Immunoglobulin Superfamily Lectins) are cell surface signaling receptors of the I-type lectin group that recognize sialic acid-bearing glycans. CD33-related-Siglecs are a subset with expression primarily in cells of hematopoietic origin and functional relevance to immune reactions. Earlier we reported a human-specific gene conversion event that markedly changed the coding region for the extracellular domain of Siglec-11, associated with human-specific expression in microglia (Hayakawa T, Angata T, Lewis AL, Mikkelsen TS, Varki NM, Varki A. 2005. A human-specific gene in microglia. Science. 309:1693). Analyzing human gene microarrays to define new patterns of expression, we observed high levels of SIGLEC11 transcript in the ovary and adrenal cortex. Thus, we examined human and chimpanzee tissues using a well-characterized anti-Siglec-11 mouse monoclonal antibody. Although adrenal expression was variable and confined to infiltrating macrophages in capillaries, ovarian expression of Siglec-11 in both humans and chimpanzees was on fibroblasts, the first example of Siglec expression on mesenchyme-derived stromal cells. Cytokines from such ovarian stromal fibroblasts play important roles in follicle development and ovulation. Stable transfection of SIGLEC11 into a primary human ovarian stromal fibroblast cell line altered the secretion of growth-regulated oncogene α, interleukin (IL)-10, IL-7, transforming growth factor β1 and tumor necrosis factor-α, cytokines involved in ovarian physiology. Probing for Siglec-11 ligands revealed distinct and strong mast cell expression in human ovaries, contrasting to diffuse stromal ligands in chimpanzee ovaries. Interestingly, there was a trend of increased Siglec-11 expression in post-menopausal ovaries compared with pre-menopausal ones. Siglec-11 expression was also found on human ovarian stromal tumors and in polycystic ovarian syndrome, a human-specific disease. These results indicate potential roles for Siglec-11 in ovarian physiology and human evolution.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Center for Academic Research and Training in Anthropogeny, Department of Medicine and Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
McLean CY, Reno PL, Pollen AA, Bassan AI, Capellini TD, Guenther C, Indjeian VB, Lim X, Menke DB, Schaar BT, Wenger AM, Bejerano G, Kingsley DM. Human-specific loss of regulatory DNA and the evolution of human-specific traits. Nature 2011; 471:216-9. [PMID: 21390129 PMCID: PMC3071156 DOI: 10.1038/nature09774] [Citation(s) in RCA: 339] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Accepted: 12/22/2010] [Indexed: 01/03/2023]
Abstract
Humans differ from other animals in many aspects of anatomy, physiology, and behaviour; however, the genotypic basis of most human-specific traits remains unknown. Recent whole-genome comparisons have made it possible to identify genes with elevated rates of amino acid change or divergent expression in humans, and non-coding sequences with accelerated base pair changes. Regulatory alterations may be particularly likely to produce phenotypic effects while preserving viability, and are known to underlie interesting evolutionary differences in other species. Here we identify molecular events particularly likely to produce significant regulatory changes in humans: complete deletion of sequences otherwise highly conserved between chimpanzees and other mammals. We confirm 510 such deletions in humans, which fall almost exclusively in non-coding regions and are enriched near genes involved in steroid hormone signalling and neural function. One deletion removes a sensory vibrissae and penile spine enhancer from the human androgen receptor (AR) gene, a molecular change correlated with anatomical loss of androgen-dependent sensory vibrissae and penile spines in the human lineage. Another deletion removes a forebrain subventricular zone enhancer near the tumour suppressor gene growth arrest and DNA-damage-inducible, gamma (GADD45G), a loss correlated with expansion of specific brain regions in humans. Deletions of tissue-specific enhancers may thus accompany both loss and gain traits in the human lineage, and provide specific examples of the kinds of regulatory alterations and inactivation events long proposed to have an important role in human evolutionary divergence.
Collapse
Affiliation(s)
- Cory Y. McLean
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Philip L. Reno
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alex A. Pollen
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Abraham I. Bassan
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Terence D. Capellini
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Catherine Guenther
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vahan B. Indjeian
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinhong Lim
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Douglas B. Menke
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bruce T. Schaar
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Aaron M. Wenger
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| | - Gill Bejerano
- Department of Computer Science, Stanford University, Stanford, CA 94305, USA
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David M. Kingsley
- Department of Developmental Biology,Stanford University School of Medicine, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
134
|
Karasik D. How pleiotropic genetics of the musculoskeletal system can inform genomics and phenomics of aging. AGE (DORDRECHT, NETHERLANDS) 2011; 33:49-62. [PMID: 20596786 PMCID: PMC3063644 DOI: 10.1007/s11357-010-9159-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Accepted: 06/14/2010] [Indexed: 04/16/2023]
Abstract
Genetic study can provide insight into the biologic mechanisms underlying inter-individual differences in susceptibility to (or resistance to) organisms' aging. Recent advances in molecular genetics and genetic epidemiology provide the necessary tools to perform a study of the genetic sources of biological aging. However, to be successful, the genetic study of a complex condition requires a heritable phenotype to be developed and validated. Genome-wide association studies offer an unbiased approach to identify new candidate genes for human diseases. It is hypothesized that convergent results from multiple aging-related traits will point out the genes responsible for the general aging of the organism. This perspective focuses on the musculoskeletal aging as an example of an approach to identify a downstream common pathway that summarizes aging processes. Since the musculoskeletal traits are linked to the state of many vital functions, disability, and ultimately survival rates, we postulate that there is significance in studying musculoskeletal aging. Construction of an integrated phenotype of aging can be achieved based on shared genetics among multiple musculoskeletal biomarkers. Valid biomarkers from other systems of the organism should be similarly explored. The new composite aging score needs to be validated by determining whether it predicts all-cause mortality, incidences of major chronic diseases, and disability late in life. Comprehensive databases on biomarkers of musculoskeletal aging in multiple large cohort studies, along with information on various health outcomes, are needed to validate the proposed measure of biological aging.
Collapse
Affiliation(s)
- David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Harvard Medical School, 1200 Centre Street, Boston, MA 02131, USA.
| |
Collapse
|
135
|
Carter AM. Comparative studies of placentation and immunology in non-human primates suggest a scenario for the evolution of deep trophoblast invasion and an explanation for human pregnancy disorders. Reproduction 2011; 141:391-6. [PMID: 21273370 DOI: 10.1530/rep-10-0530] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Deep trophoblast invasion in the placental bed has been considered the hallmark of human pregnancy. It occurs by two routes, interstitial and endovascular, and results in transformation of the walls of the spiral arteries as they traverse the decidua and the inner third of the myometrium. Disturbances in this process are associated with reproductive disorders such preeclampsia. In contrast, trophoblast invasion in Old World monkeys occurs only by the endovascular route and seldom reaches the myometrium. Recently, it was shown that this pattern is maintained in gibbons, but that the human arrangement also occurs in chimpanzee and gorilla. There is an interesting parallel with results from placental immunology regarding the evolution of the major histocompatability complex class I antigen HLA-C and its cognate receptors. HLA-C is not present in Old World monkeys or gibbons. It emerged in the orangutan and became polymorphic in the lineage leading to gorilla, bonobo, chimpanzee, and human. Interaction between HLA-C1 and HLA-C2 on the surface of trophoblast and killer immunoglobulin-like receptors (KIRs) expressed by uterine natural killer cells are important regulators of trophoblast invasion. Evolution of this system in great apes may have been one prerequisite for deep trophoblast invasion but seems to have come at a price. The evidence now suggests that certain combinations of maternal genotype for KIRs and fetal genotype for HLA-C imply an increased risk of preeclampsia, fetal growth restriction, and recurrent abortion. The fetal genotype is in part derived from the father providing an explanation for the paternal contribution to reproductive disorders.
Collapse
Affiliation(s)
- Anthony M Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, J. B. Winsloewsvej 21, DK-5000 Odense, Denmark.
| |
Collapse
|
136
|
Abstract
Overactive bladder syndrome (OAB) is a symptom-based diagnosis characterised by the presence of urinary urgency. It is highly prevalent and overlaps with the presence of bladder contractions during urine storage, which characterises the urodynamic diagnosis of detrusor overactivity. Animal models are needed to understand the pathophysiology of OAB, but the subjective nature of the symptom complex means that interpretation of the findings in animals requires caution. Because urinary urgency cannot be ascertained in animals, surrogate markers such as frequency, altered toileting areas, and non-micturition contractions have to be used instead. No model can recapitulate the subjective, objective, and related factors seen in the clinical setting. Models used include partial bladder outlet obstruction, the spontaneous hypertensive rat, the hyperlipidaemic rat, various neurological insults and some gene knock-outs. Strengths and weaknesses of these models are discussed in the context of the inherent difficulties of extrapolating subjective symptoms in animals.
Collapse
Affiliation(s)
- Brian A Parsons
- Bristol Urological Institute, Southmead Hospital, Bristol, BS10 5NB, UK.
| | | |
Collapse
|
137
|
Liu S, Lin L, Jiang P, Wang D, Xing Y. A comparison of RNA-Seq and high-density exon array for detecting differential gene expression between closely related species. Nucleic Acids Res 2011; 39:578-88. [PMID: 20864445 PMCID: PMC3025565 DOI: 10.1093/nar/gkq817] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Revised: 08/17/2010] [Accepted: 09/01/2010] [Indexed: 12/17/2022] Open
Abstract
RNA-Seq has emerged as a revolutionary technology for transcriptome analysis. In this article, we report a systematic comparison of RNA-Seq and high-density exon array for detecting differential gene expression between closely related species. On a panel of human/chimpanzee/rhesus cerebellum RNA samples previously examined by the high-density human exon junction array (HJAY) and real-time qPCR, we generated 48.68 million RNA-Seq reads. Our results indicate that RNA-Seq has significantly improved gene coverage and increased sensitivity for differentially expressed genes compared with the high-density HJAY array. Meanwhile, we observed a systematic increase in the RNA-Seq error rate for lowly expressed genes. Specifically, between-species DEGs detected by array/qPCR but missed by RNA-Seq were characterized by relatively low expression levels, as indicated by lower RNA-Seq read counts, lower HJAY array expression indices and higher qPCR raw cycle threshold values. Furthermore, this issue was not unique to between-species comparisons of gene expression. In the RNA-Seq analysis of MicroArray Quality Control human reference RNA samples with extensive qPCR data, we also observed an increase in both the false-negative rate and the false-positive rate for lowly expressed genes. These findings have important implications for the design and data interpretation of RNA-Seq studies on gene expression differences between and within species.
Collapse
Affiliation(s)
- Song Liu
- Department of Biostatistics, Roswell Park Cancer Institute, The State University of New York at Buffalo, Buffalo, NY 14203, Department of Internal Medicine and Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Lan Lin
- Department of Biostatistics, Roswell Park Cancer Institute, The State University of New York at Buffalo, Buffalo, NY 14203, Department of Internal Medicine and Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Peng Jiang
- Department of Biostatistics, Roswell Park Cancer Institute, The State University of New York at Buffalo, Buffalo, NY 14203, Department of Internal Medicine and Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Dan Wang
- Department of Biostatistics, Roswell Park Cancer Institute, The State University of New York at Buffalo, Buffalo, NY 14203, Department of Internal Medicine and Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Yi Xing
- Department of Biostatistics, Roswell Park Cancer Institute, The State University of New York at Buffalo, Buffalo, NY 14203, Department of Internal Medicine and Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
138
|
Barreiro LB, Marioni JC, Blekhman R, Stephens M, Gilad Y. Functional comparison of innate immune signaling pathways in primates. PLoS Genet 2010; 6:e1001249. [PMID: 21187902 PMCID: PMC3002988 DOI: 10.1371/journal.pgen.1001249] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 11/16/2010] [Indexed: 01/08/2023] Open
Abstract
Humans respond differently than other primates to a large number of infections. Differences in susceptibility to infectious agents between humans and other primates are probably due to inter-species differences in immune response to infection. Consistent with that notion, genes involved in immunity-related processes are strongly enriched among recent targets of positive selection in primates, suggesting that immune responses evolve rapidly, yet providing only indirect evidence for possible inter-species functional differences. To directly compare immune responses among primates, we stimulated primary monocytes from humans, chimpanzees, and rhesus macaques with lipopolysaccharide (LPS) and studied the ensuing time-course regulatory responses. We find that, while the universal Toll-like receptor response is mostly conserved across primates, the regulatory response associated with viral infections is often lineage-specific, probably reflecting rapid host–virus mutual adaptation cycles. Additionally, human-specific immune responses are enriched for genes involved in apoptosis, as well as for genes associated with cancer and with susceptibility to infectious diseases or immune-related disorders. Finally, we find that chimpanzee-specific immune signaling pathways are enriched for HIV–interacting genes. Put together, our observations lend strong support to the notion that lineage-specific immune responses may help explain known inter-species differences in susceptibility to infectious diseases. We know of a large number of diseases or medical conditions that affect humans more severely than non-human primates, such as AIDS, malaria, hepatitis B, and cancer. These differences likely arise from different immune responses to infection among species. However, due to the lack of comparative functional data across species, it remains unclear how the immune system of humans and other primates differ. In this work, we present the first genome-wide characterization of functional differences in innate immune responses between humans and our closest evolutionary relatives. Our results indicate that “core” immune responses, those that are critical to fight any invading pathogen, are the most conserved across primates and that much of the divergence in immune responses is observed in genes that are involved in response to specific microbial and viral agents. In addition, we show that human-specific immune responses are enriched for genes involved in apoptosis and cancer biology, as well as with genes previously associated with susceptibility to infectious diseases or immune-related disorders. Finally, we find that chimpanzee-specific immune signaling pathways are enriched for HIV–interacting genes. Our observations may therefore help explain known inter-species differences in susceptibility to infectious diseases.
Collapse
Affiliation(s)
- Luis B. Barreiro
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (LBB); (YG)
| | - John C. Marioni
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| | - Ran Blekhman
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
| | - Matthew Stephens
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
- Department of Statistics, University of Chicago, Chicago, Illinois, United States of America
| | - Yoav Gilad
- Department of Human Genetics, University of Chicago, Chicago, Illinois, United States of America
- * E-mail: (LBB); (YG)
| |
Collapse
|
139
|
Paar V, Glunčić M, Basar I, Rosandić M, Paar P, Cvitković M. Large Tandem, Higher Order Repeats and Regularly Dispersed Repeat Units Contribute Substantially to Divergence Between Human and Chimpanzee Y Chromosomes. J Mol Evol 2010; 72:34-55. [DOI: 10.1007/s00239-010-9401-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2010] [Accepted: 10/25/2010] [Indexed: 10/18/2022]
|
140
|
NGcGM3 ganglioside: a privileged target for cancer vaccines. Clin Dev Immunol 2010; 2010:814397. [PMID: 21048926 PMCID: PMC2965427 DOI: 10.1155/2010/814397] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 09/24/2010] [Indexed: 01/05/2023]
Abstract
Active specific immunotherapy is a promising field in cancer research. N-glycolyl (NGc) gangliosides, and particularly NGcGM3, have received attention as a privileged target for cancer therapy. Many clinical trials have been performed with the anti-NGc-containing gangliosides anti-idiotype monoclonal antibody racotumomab (formerly known as 1E10) and the conjugated NGcGM3/VSSP vaccine for immunotherapy of melanoma, breast, and lung cancer. The present paper examines the role of NGc-gangliosides in tumor biology as well as the available preclinical and clinical data on these vaccine products. A brief discussion on the relevance of prioritization of cancer antigens in vaccine development is also included.
Collapse
|
141
|
Coors ME, Glover JJ, Juengst ET, Sikela JM. The ethics of using transgenic non-human primates to study what makes us human. Nat Rev Genet 2010; 11:658-62. [PMID: 20717156 PMCID: PMC2995325 DOI: 10.1038/nrg2864] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A flood of comparative genomic data is resulting in the identification of human lineage-specific (HLS) sequences. As apes are our closest evolutionary relatives, transgenic introduction of HLS sequences into these species has the greatest potential to produce 'humanized' phenotypes and also to illuminate the functions of these sequences. We argue that such transgenic apes would also be more likely than other species to experience harm from such research, which renders such studies ethically unacceptable in apes and justifies regulatory barriers between these species and other non-human primates for HLS transgenic research.
Collapse
Affiliation(s)
- Marilyn E Coors
- Department of Psychiatry and Center for Bioethics and Humanities, School of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado 80045, USA.
| | | | | | | |
Collapse
|
142
|
Schrodi SJ. Genome-wide association scan in psoriasis: new insights into chronic inflammatory disease. Expert Rev Clin Immunol 2010; 4:565-71. [PMID: 20476959 DOI: 10.1586/1744666x.4.5.565] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Evaluation of: Liu Y, Helms C, Liao W et al. A genome-wide association study of psoriasis and psoriatic arthritis identifies new disease loci. PLoS Genet. 4, e1000041 (2008). Genome-wide association scans have delivered on their promise of revealing susceptibility polymorphisms underlying common diseases. This comprehensive psoriasis study by Liu and colleagues reports confirmation of previously identified genes (HLA-C, IL12B and IL23R), identifies several novel psoriasis loci and is the first to report psoriatic arthritis association on a genome-wide scale. Along with other recent studies, this work gives further evidence that IL-23-mediated signaling is a key component of both psoriasis and psoriatic arthritis pathogenesis. Importantly, this study provides evidence of a single-nucleotide polymorphism (SNP), 35 kb upstream of HLA-C, which is stronger than Cw 0602 - the variant traditionally attributed to the MHC-linked psoriasis-susceptibility effect. Within this region, the authors also discovered an independent SNP with very strong predisposing effects. SNPs in the COG6 region and the USP8-TNFAIP8l3 region are among the novel psoriasis associations reported. In addition, a region showing linkage on chromosome 1q demonstrated association in the epidermal differentiation complex. Four SNPs over a 439-kb region on chromosome 4q27, where KIAA1109, ADAD1 and two cytokine-encoding genes (IL2 and IL21) reside, exhibit intriguing correlation with psoriatic arthritis, although the signal strength is moderate. These results, while still preliminary, may substantially expand our knowledge of psoriasis and psoriatic arthritis genetics, opening new avenues of chronic inflammatory disease research.
Collapse
Affiliation(s)
- Steven J Schrodi
- Statistical Genetics Group, Celera, Inc., 1401 Harbor Bay Parkway, Alameda, CA 94502, USA.
| |
Collapse
|
143
|
Wu DD, Jin W, Hao XD, Tang NLS, Zhang YP. Evidence for positive selection on the Osteogenin (BMP3) gene in human populations. PLoS One 2010; 5:e10959. [PMID: 20532035 PMCID: PMC2881034 DOI: 10.1371/journal.pone.0010959] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 05/10/2010] [Indexed: 01/18/2023] Open
Abstract
Background Human skeletal system has evolved rapidly since the dispersal of modern humans from Africa, potentially driven by selection and adaptation. Osteogenin (BMP3) plays an important role in skeletal development and bone osteogenesis as an antagonist of the osteogenic bone morphogenetic proteins, and negatively regulates bone mineral density. Methodology/Principal Findings Here, we resequenced the BMP3 gene from individuals in four geographically separated modern human populations. Features supportive of positive selection in the BMP3 gene were found including the presence of an excess of nonsynonymous mutations in modern humans, and a significantly lower genetic diversity that deviates from neutrality. The prevalent haplotypes of the first exon region in Europeans demonstrated features of long-range haplotype homogeneity. In contrast with findings in European, the derived allele SNP Arg192Gln shows higher extended haplotype homozygosity in East Asian. The worldwide allele frequency distribution of SNP shows not only a high-derived allele frequency in Asians, but also in Americans, which is suggestive of functional adaptation. Conclusions/Significance In conclusion, we provide evidence for recent positive selection operating upon a crucial gene in skeletal development, which may provide new insight into the evolution of the skeletal system and bone development.
Collapse
Affiliation(s)
- Dong-Dong Wu
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Graduate School of the Chinese Academy of Sciences, Beijing, China
| | - Wei Jin
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
| | - Xiao-Dan Hao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Graduate School of the Chinese Academy of Sciences, Beijing, China
| | - Nelson Leung Sang Tang
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China
- Department of Chemical Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
- Laboratory for Genetics of Disease Susceptibility, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Laboratory for Conservation and Utilization of Bio-resource, Yunnan University, Kunming, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China
- * E-mail:
| |
Collapse
|
144
|
Colloquium paper: uniquely human evolution of sialic acid genetics and biology. Proc Natl Acad Sci U S A 2010; 107 Suppl 2:8939-46. [PMID: 20445087 DOI: 10.1073/pnas.0914634107] [Citation(s) in RCA: 206] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Darwinian evolution of humans from our common ancestors with nonhuman primates involved many gene-environment interactions at the population level, and the resulting human-specific genetic changes must contribute to the "Human Condition." Recent data indicate that the biology of sialic acids (which directly involves less than 60 genes) shows more than 10 uniquely human genetic changes in comparison with our closest evolutionary relatives. Known outcomes are tissue-specific changes in abundant cell-surface glycans, changes in specificity and/or expression of multiple proteins that recognize these glycans, and novel pathogen regimes. Specific events include Alu-mediated inactivation of the CMAH gene, resulting in loss of synthesis of the Sia N-glycolylneuraminic acid (Neu5Gc) and increase in expression of the precursor N-acetylneuraminic acid (Neu5Ac); increased expression of alpha2-6-linked Sias (likely because of changed expression of ST6GALI); and multiple changes in SIGLEC genes encoding Sia-recognizing Ig-like lectins (Siglecs). The last includes binding specificity changes (in Siglecs -5, -7, -9, -11, and -12); expression pattern changes (in Siglecs -1, -5, -6, and -11); gene conversion (SIGLEC11); and deletion or pseudogenization (SIGLEC13, SIGLEC14, and SIGLEC16). A nongenetic outcome of the CMAH mutation is human metabolic incorporation of foreign dietary Neu5Gc, in the face of circulating anti-Neu5Gc antibodies, generating a novel "xeno-auto-antigen" situation. Taken together, these data suggest that both the genes associated with Sia biology and the related impacts of the environment comprise a relative "hot spot" of genetic and physiological changes in human evolution, with implications for uniquely human features both in health and disease.
Collapse
|
145
|
Soto PC, Stein LL, Hurtado-Ziola N, Hedrick SM, Varki A. Relative over-reactivity of human versus chimpanzee lymphocytes: implications for the human diseases associated with immune activation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 184:4185-95. [PMID: 20231688 PMCID: PMC3085894 DOI: 10.4049/jimmunol.0903420] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Although humans and chimpanzees share >99% identity in alignable protein sequences, they differ surprisingly in the incidence and severity of some common diseases. In general, humans infected with various viruses, such as HIV and hepatitis C virus, appear to develop stronger reactions and long-term complications. Humans also appear to suffer more from other diseases associated with over-reactivity of the adaptive immune system, such as asthma, psoriasis, and rheumatoid arthritis. In this study, we show that human T cells are more reactive than chimpanzee T cells to a wide variety of stimuli, including anti-TCR Abs of multiple isotypes, l-phytohemagglutin, Staphylococcus aureus superantigen, a superagonist anti-CD28 Ab, and in MLRs. We also extend this observation to B cells, again showing a human propensity to react more strongly to stimuli. Finally, we show a relative increase in activation markers and cytokine production in human lymphocytes in response to uridine-rich (viral-like) ssRNA. Thus, humans manifest a generalized lymphocyte over-reactivity relative to chimpanzees, a finding that is correlated with decreased levels of inhibitory sialic acid-recognizing Ig-superfamily lectins (Siglecs; particularly Siglec-5) on human T and B cells. Furthermore, Siglec-5 levels are upregulated by activation in chimpanzee but not human lymphocytes, and human T cell reactivity can be downmodulated by forced expression of Siglec-5. Thus, a key difference in the immune reactivity of chimp and human lymphocytes appears to be related to the differential expression of Siglec-5. Taken together, these data may help explain human propensities for diseases associated with excessive activation of the adaptive immune system.
Collapse
MESH Headings
- Acquired Immunodeficiency Syndrome/immunology
- Acquired Immunodeficiency Syndrome/metabolism
- Acquired Immunodeficiency Syndrome/pathology
- Adaptive Immunity/genetics
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/physiology
- Antigens, Differentiation, Myelomonocytic/biosynthesis
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/physiology
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- B-Lymphocytes/virology
- Cell Proliferation
- Cells, Cultured
- Down-Regulation/genetics
- Down-Regulation/immunology
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/genetics
- Growth Inhibitors/physiology
- HIV Infections/immunology
- HIV Infections/metabolism
- HIV Infections/pathology
- Hepatitis C/immunology
- Hepatitis C/metabolism
- Hepatitis C/pathology
- Humans
- Lectins/biosynthesis
- Lectins/genetics
- Lectins/physiology
- Lymphocyte Activation/immunology
- Pan troglodytes/immunology
- Receptors, Cell Surface/physiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/virology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Paula C. Soto
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Lance L. Stein
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Nancy Hurtado-Ziola
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Stephen M. Hedrick
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Ajit Varki
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
146
|
Anbazhagan P, Purushottam M, Kiran Kumar HB, Mukherjee O, Jain S, Sowdhamini R. Phylogenetic Analysis and Selection Pressures of 5-HT Receptors in Human and Non-human Primates: Receptor of an Ancient Neurotransmitter. J Biomol Struct Dyn 2010; 27:581-98. [DOI: 10.1080/07391102.2010.10508573] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
147
|
A human-specific de novo protein-coding gene associated with human brain functions. PLoS Comput Biol 2010; 6:e1000734. [PMID: 20376170 PMCID: PMC2845654 DOI: 10.1371/journal.pcbi.1000734] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 03/03/2010] [Indexed: 02/05/2023] Open
Abstract
To understand whether any human-specific new genes may be associated with human brain functions, we computationally screened the genetic vulnerable factors identified through Genome-Wide Association Studies and linkage analyses of nicotine addiction and found one human-specific de novo protein-coding gene, FLJ33706 (alternative gene symbol C20orf203). Cross-species analysis revealed interesting evolutionary paths of how this gene had originated from noncoding DNA sequences: insertion of repeat elements especially Alu contributed to the formation of the first coding exon and six standard splice junctions on the branch leading to humans and chimpanzees, and two subsequent substitutions in the human lineage escaped two stop codons and created an open reading frame of 194 amino acids. We experimentally verified FLJ33706's mRNA and protein expression in the brain. Real-Time PCR in multiple tissues demonstrated that FLJ33706 was most abundantly expressed in brain. Human polymorphism data suggested that FLJ33706 encodes a protein under purifying selection. A specifically designed antibody detected its protein expression across human cortex, cerebellum and midbrain. Immunohistochemistry study in normal human brain cortex revealed the localization of FLJ33706 protein in neurons. Elevated expressions of FLJ33706 were detected in Alzheimer's brain samples, suggesting the role of this novel gene in human-specific pathogenesis of Alzheimer's disease. FLJ33706 provided the strongest evidence so far that human-specific de novo genes can have protein-coding potential and differential protein expression, and be involved in human brain functions.
Collapse
|
148
|
Kelley DR, Salzberg SL. Detection and correction of false segmental duplications caused by genome mis-assembly. Genome Biol 2010; 11:R28. [PMID: 20219098 PMCID: PMC2864568 DOI: 10.1186/gb-2010-11-3-r28] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Revised: 12/11/2009] [Accepted: 03/10/2010] [Indexed: 11/23/2022] Open
Abstract
Diploid genomes with divergent chromosomes present special problems for assembly software as two copies of especially polymorphic regions may be mistakenly constructed, creating the appearance of a recent segmental duplication. We developed a method for identifying such false duplications and applied it to four vertebrate genomes. For each genome, we corrected mis-assemblies, improved estimates of the amount of duplicated sequence, and recovered polymorphisms between the sequenced chromosomes.
Collapse
Affiliation(s)
- David R Kelley
- Center for Bioinformatics and Computational Biology, Institute for Advanced Computer Studies, University of Maryland, College Park, MD 20742, USA
| | - Steven L Salzberg
- Center for Bioinformatics and Computational Biology, Institute for Advanced Computer Studies, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
149
|
Nowick K, Stubbs L. Lineage-specific transcription factors and the evolution of gene regulatory networks. Brief Funct Genomics 2010; 9:65-78. [PMID: 20081217 DOI: 10.1093/bfgp/elp056] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nature is replete with examples of diverse cell types, tissues and body plans, forming very different creatures from genomes with similar gene complements. However, while the genes and the structures of proteins they encode can be highly conserved, the production of those proteins in specific cell types and at specific developmental time points might differ considerably between species. A full understanding of the factors that orchestrate gene expression will be essential to fully understand evolutionary variety. Transcription factor (TF) proteins, which form gene regulatory networks (GRNs) to act in cooperative or competitive partnerships to regulate gene expression, are key components of these unique regulatory programs. Although many TFs are conserved in structure and function, certain classes of TFs display extensive levels of species diversity. In this review, we highlight families of TFs that have expanded through gene duplication events to create species-unique repertoires in different evolutionary lineages. We discuss how the hierarchical structures of GRNs allow for flexible small to large-scale phenotypic changes. We survey evidence that explains how newly evolved TFs may be integrated into an existing GRN and how molecular changes in TFs might impact the GRNs. Finally, we review examples of traits that evolved due to lineage-specific TFs and species differences in GRNs.
Collapse
Affiliation(s)
- Katja Nowick
- Department of Cell and Developmental Biology, Institute for Genomic Biology, University of Illinois, 1206 W. Gregory Drive, Urbana, IL 61802, USA
| | | |
Collapse
|
150
|
Abstract
Advocates of chimpanzee research claim the genetic similarity of humans and chimpanzees make them an indispensable research tool to combat human diseases. Given that cancer is a leading cause of human death worldwide, one might expect that if chimpanzees were needed for, or were productive in, cancer research, then they would have been widely used. This comprehensive literature analysis reveals that chimpanzees have scarcely been used in any form of cancer research, and that chimpanzee tumours are extremely rare and biologically different from human cancers. Often, chimpanzee citations described peripheral use of chimpanzee cells and genetic material in predominantly human genomic studies. Papers describing potential new cancer therapies noted significant concerns regarding the chimpanzee model. Other studies described interventions that have not been pursued clinically. Finally, available evidence indicates that chimpanzees are not essential in the development of therapeutic monoclonal antibodies. It would therefore be unscientific to claim that chimpanzees are vital to cancer research. On the contrary, it is reasonable to conclude that cancer research would not suffer, if the use of chimpanzees for this purpose were prohibited in the US. Genetic differences between humans and chimpanzees, make them an unsuitable model for cancer, as well as other human diseases.
Collapse
Affiliation(s)
- Jarrod Bailey
- New England Anti-Vivisection Society, Boston, MA 02108-5100, USA.
| |
Collapse
|