With world population increase, the issue of created food became the global big problem. With the population increase such as China and India, Afro-Asian countries, the frequent occurrence of the international dispute by the intensification of the struggle, the acquisition competition of the mineral resources, food resources and the industrial use begins to affect our life not expectation practically.
We must build the world of the peace with the hope for our posterity. Development of the field of industry that is new in correspondence with world population increase and industrial labor market, employment, natural reproduction circulatory system (we must build economy, the social system of sustainable).
we developed agriculture, stock raising for food which used the utilization of the sea area that occupied approximately 70% of surface area of the earth and the use and the marine ranches such as marine resources (sea life, bottom of the sea mineral), large-scale floating form ocean structure, and Itbecameinorderoftheworldpeaceinthetimeswhenitwasnecessarytotrytobeabletolivewiththetranquility.
For example, we bring up upbringing of new industry such as the marine wind-generated electricity, ocean photovoltaic power generation and new industry, and, for the purpose of new job creation, a grows new marine resources development business, industry, and promotion develops the marine resources development-affiliated new business, project based on the idea of the firm peace, and go ahead through the international cooperation while opening a dream and the hope of the world young man, and tranquility and hope will make the future society which there is.
Zakir Naik is an Indian public speaker on the subject of Islam and comparative religion. He is the founder and president of the Islamic Research Foundation (IRF),[1][2] He is sometimes referred to as a televangelist because of his work at Peace TV.[3][4] Before becoming a public speaker, he trained as a medical doctor.[4] He has published booklet versions of lectures on Islam and comparative religion. Although he has publicly disclaimed sectarianism in Islam, he is regarded as an exponent of the Salafi ideology.[5][6][7] Biography
Zakir Naik was born in Mumbai, Maharastra, India. He attended St. Peter's High School in Mumbai. Later he enrolled at Kishinchand Chellaram College, before studying medicine at Topiwala National Medical College and Nair Hospital and later the University of Mumbai, where he obtained a Bachelor of Medicine and Surgery (MBBS).[1][non-primary source needed] His wife, Farhat Naik, works for the women's section of the IRF.[8]
In 1991 he started working in the field of Dawah, and founded the IRF.[9] Naik says he was inspired by Ahmed Deedat, an Islamic preacher, having met him in 1987.[10] (Naik is sometimes referred to as "Deedat plus", a label given to him by Deedat himself.)[10][11] Naik says that his goal is to "concentrate on the educated Muslim youth who have become apologetic about their own religion and have started to feel the religion is outdated".[12] He considers it a duty of every Muslim to remove perceived misconceptions about Islam and to counter what he views as the Western media's anti-Islamic bias in the aftermath of the September 11, 2001 attacks in the United States.[13] Naik has said that "despite the strident anti-Islam campaign, 34,000 Americans have embraced Islam from September 2001 to July 2002". He says Islam is a religion of reason and logic, and that the Quran contains 1000 verses relating to science, which he says explains the number of Western converts.[14] Some of his articles are published in magazines such as Islamic Voice.[15]
Naik is the founder of the Islamic International School in Mumbai.[16] and United Islamic Aid, which provides scholarship to poor and destitute Muslim youth.[17] Lectures and debates
Naik has held many debates and lectures around the world. Anthropologist Thomas Blom Hansen has written that Naik's style of memorising the Quran and Hadith literature in various languages, and his related missionary activity, has made him extremely popular in Muslim circles.[12] Many of his debates are recorded and widely distributed in video and DVD media and online. His talks are usually recorded in English and broadcast on weekends on several cable networks in Mumbai's Muslim neighbourhoods, and on the Peace TV channel, which he co-produces.[18][19] Topics he speaks on include: "Islam and Modern Science", "Islam and Christianity", and "Islam and secularism".
One of Naik's most-cited debates was with William Campbell in Chicago in April 2000, on the topic of "The Qur'an and the Bible: In the Light of Science".[20] On 21 January 2006 Naik held an inter-religious dialogue with Sri Sri Ravi Shankar in Bangalore about the concept of God in Islam and Hinduism.[21] In February 2011 Naik addressed the Oxford Union via video link from India.[22] Every year since November 2007 Naik has led a 10-day Peace Conference at Somaiya Ground, Sion, Mumbai. Lectures on Islam have been presented by Naik and twenty other Islamic speakers.[23]
Naik argues that scientific theories were prophesised by the Quran. For example, he says certain verses of the Quran accurately describe embryological development.[24] Recognition Islamic Personality of the Year Award 2013 from The Dubai International Holy Quran Award.[25][26] The award was presented by Hamdan bin Rashid Al Maktoum, Ruler of Dubai and Minister of Finance and Industry of the United Arab Emirates.[27]
On 5 November 2013, the Department of Islamic Development Malaysia conferred a Ma'al Hijrah Distinguished Personality award to Naik.[28] In a ceremony at the Putrajaya International Convention Centre, the award was presented by Yang di-Pertuan Agong, Malaysia's head of state.
Views Biological evolution
Naik has said that the theory of evolution is "only a hypothesis, and an unproven conjecture at best".[30] According to Naik, most scientists "support the theory, because it went against the Bible – not because it was true."[31] Apostasy
Naik has said that not all Muslims who convert from Islam should necessarily receive death sentences, but that those who leave Islam and then "propagate the non-Islamic faith and speak against Islam" should be put to death in an Islamic rule.[32][33] Terrorism
Naik's views and statements on terrorism have at times been criticised in the media. In a YouTube video, speaking of Osama bin Laden, Naik said that he would not criticise bin Laden because he had not met him and did not know him personally. He added that, "If bin Laden is fighting enemies of Islam, I am for him," and that "If he is terrorizing America – the terrorist, biggest terrorist – I am with him. Every Muslim should be a terrorist. The thing is that if he is terrorizing the terrorist, he is following Islam. Whether he is or not, I don’t know, but you as Muslims know that, without checking up, laying allegations is also wrong."[34] When Time hinted that this remark could have inspired Najibullah Zazi's terrorist activities, Naik insisted: "I have always condemned terrorism, because according to the glorious Koran, if you kill one innocent person, then you have killed the whole of humanity".[34]
In 2010, Naik said that he had been quoted out of context regarding the remarks on terrorism. "As far as terrorist is concerned," he said, "I tell the Muslims that every Muslim should be a terrorist. ... What is the meaning of the word terrorist? Terrorist by definition means a person who terrorises. So in this context every Muslim should be a terrorist to each and every anti-social element. I’m aware that terrorist is more commonly used for a person who terrorises innocent human beings. So in this context no Muslim should ever terrorise a single innocent human being."[35]
In a lecture delivered on 31 July 2008 on Peace TV, Naik commented on the attacks of 11 September: "it is a blatant, open secret that this attack on the Twin Towers was done by George Bush himself".[36] Propagation of other faiths in Islamic states
Naik says that propagation of other religions within an Islamic state is forbidden while he appreciates people of other religions allowing Muslims to freely propagate Islam in their country. Naik explains this by saying that, for example, mathematics teachers must teach that 2+2=4 and not 2+2=3 or 5. Likewise, Naik argues, “regarding building of churches or temples, how can we allow this when their religion is wrong and when their worshipping is wrong?”[37] Other countries Visit to Australia and Wales
In 2004 Naik, at the invitation of the Islamic Information and Services Network of Australasia, made an appearance at Melbourne University, where he argued that only Islam gave women true equality.[38] He said the more "revealing Western dress" makes women more susceptible to rape.[39] Sushi Das of The Age commented that "Naik extolled the moral and spiritual superiority of Islam and lampooned other faiths and the West in general", further criticising that Naik's words "fostered a spirit of separateness and reinforced prejudice".[40]
In August 2006 Naik's visit and conference in Cardiff caused controversy when Welsh MPDavid Davies called for his appearance to be cancelled. He said Naik was a "hate-monger", and that his views did not deserve a public platform; Muslims from Cardiff, however, defended Naik's right to speak in the city. Saleem Kidwai, Secretary General of the Muslim Council of Wales, disagreed with Davies, stating that "people who know about him [Naik] know that he is one of the most uncontroversial persons you could find. He talks about the similarities between religions, and how should we work on the common ground between them", whilst also inviting Davies to discuss further with Naik personally in the conference. The conference went ahead, after the Cardiff council stated it was satisfied that he would not be preaching extremist views.[41] 2010 exclusion from the UK and Canada
Naik was denied entry into the United Kingdom and Canada in June 2010.[42][43] He was banned from entering the UK by Home SecretaryTheresa May after arranging to give talks in London and Sheffield. May said of the exclusion order, "Numerous comments made by Dr Naik are evidence to me of his unacceptable behaviour".[42] Naik argued that the Home Secretary was making a political decision and not a legal one, and his lawyer said the decision was "barbaric and inhuman". He also claimed that his comments were taken out of context.[44] Film producer Mahesh Bhatt supported Naik, saying the ban constituted an attack on freedom of speech.[45] It was reported that Naik would attempt to challenge the ruling in the High Court.[46] His application for judicial review was dismissed on 5 November 2010.[9] Naik was forbidden from entering Canada after Tarek Fatah, founder of the Muslim Canadian Congress, warned MPs of Naik's views.[43] Visit to Malaysia in 2012
Naik delivered four lectures in Malaysia during 2012. The lectures took place in Johor Baru, Universiti Teknologi MARA in Shah Alam,[47]Kuantan and Putra World Trade Centre in Kuala Lumpur.The former Prime Minister of Malaysia, Mahathir Mohamad, prominent figures and several thousand people attended the lectures at different places despite protest by the members of a banned group, HINDRAF.[48] The organizers of Naik's speeches said their purpose was to promote harmony among people of various religions.[49] Reception, Awards, Titles and Honors Naik was ranked 89 on The Indian Express's list of the "100 Most Powerful Indians in 2010".[50] He was ranked 82 in the 2009 edition.[51] According to Praveen Swami, Naik is "perhaps the most influential Salafi ideologue in India".[52]Sanjiv Buttoo says he is acknowledged as an authority on Islam, but is known for making negative remarks about other religions.[42]Sadanand Dhume writes that Naik has a "carefully crafted image of moderation", because of his gentle demeanour, his wearing of a suit and tie, and his quoting of scriptures of other religions.[53] He is also listed in the book "The 500 Most Influential Muslims" under honourable mention, in the 2009,[54] 2010, 2011, 2012 and 2013/2014 [55] editions.[56] In July 2013, Naik was named as the Islamic Personality of the Year, announced by the 17th Dubai International Holy Quran Award (DIHQA).[57][58]
Criticism In The Wall Street Journal, Sadanand Dhume criticised Naik for recommending the death penalty for homosexuals and for apostasy from the faith.[62] He also criticised him for calling for India to be ruled by Shariah law. He added that, according to Naik, Jews "control America" and are the "strongest in enmity to Muslims." He maintained that Naik supports a ban on the construction of non-Muslim places of worship in Muslim lands as well as the Taliban's bombing of the Bamiyan Buddhas. Dhume argues that people reportedly drawn to Naik's message include Najibullah Zazi, the Afghan-American arrested for planning suicide attacks on the New York subway; Rahil Sheikh, accused of involvement in a series of train bombings in Bombay in 2006; and Kafeel Ahmed, the Bangalore man fatally injured in a failed suicide attack on Glasgow airport in 2007. He concluded that unless Indians find the ability to criticise such a radical Islamic preacher as robustly as they would a Hindu equivalent, the idea of Indian secularism would remain deeply flawed.[62] The Times of India published a profile of Naik entitled "The controversial preacher" after he was banned from the United Kingdom. According to The Times, "the fact is that barring the band of Muslims whose bruised egos Naik suitably massages through his Islam supremacist talks, most rational Muslims and non-Muslims find his brand of Islam a travesty of the faith". The Times also claimed that "the Wahabi-Salafist brand of Islam, bankrolled by petro-rich Saudi Arabia and propagated by preachers like Naik, does not appreciate the idea of pluralism". The article quotes Muslim scholar Wahiduddin Khan: "Dawah, which Naik also claims to be engaged in, is to make people aware of the creation plan of God, not to peddle some provocative, dubious ideas as Naik does". He adds: "The wave of Islamophobia in the aftermath of 9/11 and the occupation of Iraq and Afghanistan have only added to the Muslims’ sense of injury. In such a situation, when a debater like Zakir Naik, in eloquent English, takes on preachers of other faiths and defeats them during debates, the Muslims’ chests puff with pride. A community nursing a huge sense of betrayal and injustice naturally lionises anyone who gives it a sense of pride. Never mind if it’s false pride".[63]
Indian journalist Khushwant Singh says he "disagree[s] with almost everything [Naik] has to say about misconceptions about Islam". Singh argues that Naik's pronouncements are "juvenile", and said "they seldom rise above the level of undergraduate college debates, where contestants vie with each other to score brownie points".[64] Singh also says Naik's audiences "listen to him with rapt attention and often explode in enthusiastic applause when he rubbishes other religious texts".[65] Torkel Brekke, a professor of religious history in Norway, calls Naik a "very controversial figure" because of his rhetorical attack on other religions and other varieties of Islam. He writes that Naik is "strongly disliked" by many members of the Indian ulema for ignoring their authority and stating that anybody can interpret the Quran.[66] Conservative Deobandi mullahs have accused Naik of "destroying Islam" by driving Muslims away from the correct religious authorities.[67] Khaled Ahmed criticised Naik for "indirectly support[ing]" Al-Qaeda by referring to Osama bin Laden as a "soldier of Islam".[68] In 2008 an Islamic scholar in Lucknow, shahar qazi Mufti Abul Irfan Mian Firangi Mahali, issued a fatwa against Naik, saying that he supported Osama bin Laden, and that his teachings were un-Islamic.[69] Praveen Swami considers Naik to be a part of the ideological infrastructure created to feed "Tempered Jihad", which he defines as Jihad calibrated to advance Islamist political objectives.[52] Swami argued that some of Naik’s teachings are similar to those of organizations advocating violence, although Naik himself emphatically rejects terrorism.[70] According to Swami, Naik's IRF has proved to be a "magnet" for figures linked to the Lashkar-e-Taiba, while his message has mesmerised violent Islamists, and his works "help make sense of the motivations of Indian recruits to the jihad."[52]
This page was last modified on 24 February 2015
During the Exodus, one of the most famous miracles of the Old Testament took place. More than 3000 years have passed since Moses led more than 2 million Israelites across the Red Sea and out of bondage to Egypt and its Pharaoh. This film follows the footsteps of Moses and the Israelites and reveals physical evidence for the Exodus including 3800 year old remains of Hebrew settlements in Egypt, Egyptian records of the Hebrew bondage in Egypt, the precise route they would have followed to freedom, their crossing site on the shore of the Red Sea, the location of Mt. Sinai, and more. Additional information on this and other topics can be found at my website: http://www.taughtbytheholyspirit.com/
The Exodus (from Greek ἔξοδος exodos, "going out") is the founding myth of Israel; its message is that the Israelites were delivered from slavery by Yahweh and therefore belong to him through the Mosaic covenant.[1] It tells of the enslavement of the Israelites in Egypt following the death of Joseph, their departure under the leadership of Moses, the revelations at Sinai, and their wanderings in the wilderness up to the borders of Canaan.[2]
The archeological evidence does not support the story told in the Book of Exodus[3] and most archaeologists have abandoned the investigation of Moses and the Exodus as "a fruitless pursuit".[4] The opinion of the overwhelming majority of modern biblical scholars is that the Pentateuch was shaped into its final form in the post-Exilic period,[5] although the traditions behind it are older and can be traced in the writings of the 8th century BCE prophets.[6] How far beyond that the tradition might stretch cannot be told: "Presumably an original Exodus story lies hidden somewhere inside all the later revisions and alterations, but centuries of transmission have long obscured its presence, and its substance, accuracy and date are now difficult to determine."[7]
The overall intent of the books of Exodus, Leviticus, Numbers and Deuteronomy was to demonstrate God's actions in history, to recall Israel's bondage and salvation, and to demonstrate the fulfillment of Israel's covenant.[7] The Exodus has been central to Judaism: it served to orient Jews towards the celebration of God's actions in history, in contrast to polytheistic celebrations of the gods' actions in nature, and even today it is recounted daily in Jewish prayers and celebrated in the festival of Pesach. In secular history the exodus has served as inspiration and model for many groups, from early Protestant settlers fleeing persecution in Europe to 19th and 20th century African-Americans striving for freedom and civil rights.[8]
Origins of the Exodus story The opinion of the overwhelming majority of modern biblical scholars is that the Torah (the series of five books which consist of the book of Genesis plus the books in which the Exodus story is told) was shaped in the post-Exilic period.[5] There are currently two important hypotheses explaining the background to this: the first is Persian Imperial authorisation, the idea that the post-Exilic community needed a legal basis on which to function within the Persian Imperial system; the second relates to the community of citizens organised around the Temple, with the Pentateuch providing the criteria for who would belong to it (the narratives and genealogies in Genesis) and establishing the power structures and relative positions of its various groups.[9] In either case, the Book of Exodus forms a "charter myth" for Israel: Israel was delivered from slavery by Yahweh and therefore belongs to him through the covenant.[1]
The completion of the Torah and its elevation to the center of post-Exilic Judaism was as much or more about combining older texts as writing new ones – the final Pentateuch was based on earlier traditions.[10] While the story in the books of Exodus, Numbers and Deuteronomy is the best-known account of the Exodus, there are over 150 references throughout the Bible.[11] The earliest mentions are in the prophets Amos (possibly) and Hosea (certainly), both active in 8th century BCE Israel; in contrast Proto-Isaiah and Micah, both active in Judah at much the same time, never do; it thus seems reasonable to conclude the Exodus tradition was important in the northern kingdom in the 8th century BCE, but not in Judah.[6]
In a recent work, Stephen C. Russell traces the 8th century BCE prophetic tradition to three originally separate variants, in the northern kingdom of Israel, in Trans-Jordan, and in the southern kingdom of Judah. Russell proposes different hypothetical historical backgrounds to each tradition: the tradition from Israel, which involves a journey from Egypt to the region of Bethel, he suggests is a memory of herders who could move to and from Egypt in times of crisis; for the Trans-Jordanian tradition, which focuses on deliverance from Egypt without a journey, he suggests a memory of the withdrawal of Egyptian control at the end of the Late Bronze Age; and for Judah, whose tradition is preserved in the Song of the Sea, he suggests the celebration of a military victory over Egypt, although it is impossible to suggest what this victory may have been.[11] Cultural significanceThe exodus is remembered daily in Jewish prayers and celebrated each year at the feast of Passover. [12] The Hebrew name for this festival, Pesach, refers to God's instruction to the Israelites to prepare unleavened bread as they would be leaving Egypt in haste, and to mark their doors with the blood of slaughtered sheep so that the "Angel" or "the destroyer" tasked with killing the first-born of Egypt would "pass over" them. (Despite the Exodus story, scholars believe that the Passover festival originated not in the biblical story but as a magic ritual to turn away demons from the household.)[13]
Jewish tradition has preserved national and personal reminders of this pivotal narrative in daily life. Examples include the wearing of tefillin (phylacteries) on the arm and forehead, the wearing of tzitzit (knotted ritual fringes attached to the four corners of the prayer shawl), the eating of matzot (unleavened bread) during the Pesach, the fasting of the firstborn a day before Pesach, and the redemption of firstborn children and animals.
Historicity Most histories of ancient Israel no longer consider information about the Exodus recoverable or even relevant to the story of Israel's emergence.[14] Nevertheless, the discussion of the historicity of the exodus has a long history, and continues to attract attention.
Numbers and logistics The consensus among biblical scholars today is that there was never any exodus of the proportions described in the Bible.[15] According to Exodus 12:37–38, the Israelites numbered "about six hundred thousand men on foot, besides women and children," plus many non-Israelites and livestock.[16]Numbers 1:46 gives a more precise total of 603,550 men aged 20 and up.[17] The 600,000, plus wives, children, the elderly, and the "mixed multitude" of non-Israelites would have numbered some 2 million people,[18] compared with an entire Egyptian population in 1250 BCE of around 3 to 3.5 million.[19] Marching ten abreast, and without accounting for livestock, they would have formed a line 150 miles long.[20] No evidence has been found that indicates Egypt ever suffered such a demographic and economic catastrophe or that the Sinai desert ever hosted (or could have hosted) these millions of people and their herds.[21]
Some scholars have rationalised these numbers into smaller figures, for example reading the Hebrew as "600 families" rather than 600,000 men, but all such solutions have their own set of problems.[22] The view of mainstream modern biblical scholarship is that the improbability of the Exodus story originates because it was written not as history, but to demonstrate God's purpose and deeds with his Chosen People, Israel.[3] Some have suggested that the 603,550 people delivered from Egypt (according to Numbers 1:46) is not a number, but a gematria (a code in which numbers represent letters or words) for bnei yisra'el kol rosh, "the children of Israel, every individual;"[23] while the number 600,000 symbolises the total destruction of the generation of Israel which left Egypt, none of whom lived to see the Promised Land.[24] Archaeology A century of research by archaeologists and Egyptologists has found no evidence which can be directly related to the Exodus captivity and the escape and travels through the wilderness,[3] and most archaeologists have abandoned the archaeological investigation of Moses and the Exodus as "a fruitless pursuit".[4] A number of theories have been put forward to account for the origins of the Israelites, and despite differing details they agree on Israel's Canaanite origins.[25] The culture of the earliest Israelite settlements is Canaanite, their cult-objects are those of the Canaanite god El, the pottery remains in the local Canaanite tradition, and the alphabet used is early Canaanite, and almost the sole marker distinguishing the "Israelite" villages from Canaanite sites is an absence of pig bones, although whether even this is an ethnic marker or is due to other factors remains a matter of dispute.[26] Anachronisms Despite the Bible's internal dating of the Exodus to the 2nd millennium BCE, details point to a 1st millennium date for the composition of the Book of Exodus: Ezion-Geber, (one of the Stations of the Exodus), for example, dates to a period between the 8th and 6th centuries BCE with possible further occupation into the 4th century BCE,[27] and those place-names on the Exodus route which have been identified – Goshen, Pithom, Succoth, Ramesses and Kadesh Barnea – point to the geography of the 1st millennium rather than the 2nd.[28] Similarly, Pharaoh's fear that the Israelites might ally themselves with foreign invaders seems unlikely in the context of the late 2nd millennium, when Canaan was part of an Egyptian empire and Egypt faced no enemies in that direction, but does make sense in a 1st millennium context, when Egypt was considerably weaker and faced invasion first from the Persians and later from Seleucid Syria.[29] The mention of the dromedary in Exodus 9:3 also suggests a later date of composition – the widespread domestication of the camel as a herd animal was thought not to have taken place before the late 2nd millennium, after the Israelites had already emerged in Canaan,[30] and they did not become widespread in Egypt until c.200–100 BCE.[31] ChronologyThe chronology of the Exodus story likewise underlines its essentially religious rather than historical nature. The number seven, for example, was sacred to God in Judaism, and so the Israelites arrive at Sinai, where they will meet God, at the beginning of the seventh week after their departure from Egypt,[32] while the erection of the Tabernacle, God's dwelling-place among his people, occurs in the year 2666 after God creates the world, two-thirds of the way through a four thousand year era which culminates in or around the re-dedication of the Second Temple in 164 BCE.[33][34][Notes 1]
Route Main article: Stations list
The Torah lists the places where the Israelites rested. A few of the names at the start of the itinerary, including Ra'amses, Pithom and Succoth, are reasonably well identified with archaeological sites on the eastern edge of the Nile delta,[28] as is Kadesh-Barnea, where the Israelites spend 38 years after turning back from Canaan, but other than that very little is certain. The crossing of the Red Sea has been variously placed at the Pelusic branch of the Nile, anywhere along the network of Bitter Lakes and smaller canals that formed a barrier toward eastward escape, the Gulf of Suez (SSE of Succoth) and the Gulf of Aqaba (S of Ezion-Geber), or even on a lagoon on the Mediterranean coast. The biblical Mt. Sinai is identified in Christian tradition with Jebel Musa in the south of the Sinai Peninsula, but this association dates only from the 3rd century CE and no evidence of the Exodus has been found there.[35]
Date Main article: Pharaoh of the Exodus
Attempts to date the Exodus to a specific century have been inconclusive.[36]1 Kings 6:1 says that the Exodus occurred 480 years before the construction of Solomon's Temple; this would imply an Exodus c.1446 BCE, during Egypt's Eighteenth Dynasty.[37] However, it is widely recognised that the number in 1 Kings is symbolic,[38] representing twelve generations of forty years each.[39] (The number 480 is not only symbolic – the twelve generations – but schematic: Solomon's temple (the First Temple) is founded 480 years after the Exodus and 480 years before the foundation of the Second Temple).[40] There are also major archeological obstacles in dating the Exodus to the Eighteenth Dynasty: Canaan at the time was a part of the Egyptian empire, so that the Israelites would in effect be escaping from Egypt to Egypt, and its cities were unwalled and do not show destruction layers consistent with the Bible's account of the occupation of the land (e.g., Jericho was "small and poor, almost insignificant, and unfortified (and) [t]here was also no sign of a destruction". (Finkelstein and Silberman, 2002).[41] William F. Albright, the leading biblical archaeologist of the mid-20th century, proposed an alternative 13th century date of around 1250–1200 BCE for the Exodus event and the entry into Canaan described in the book of Joshua.[42] (The Merneptah Stele indicated that a people called "Israel" were already known in Canaan by the reign of Merneptah (1213–1203 BCE), so a date later than this was impossible). His argument was based on many strands of evidence, including archaeologically attested destruction at Beitel (Bethel) and some other cities at around that period and the occurrence of distinctive house-types and round-collared jars which, in his opinion, were "Israelite".[42] Albright's theory enjoyed popularity at the time, but has now been generally abandoned in scholarship:[42] the so-called "Israelite" house-type, the collar-rimmed jars, and other items which Albright thought distinctive and new have now been recognised as continuations of indigenous Canaanite types,[43] and while some "Joshua" cities, including Hazor, Lachish, Megiddo and others, have destruction and transition layers around 1250–1145 BCE, others, including Jericho, have none or were uninhabited during this period.[44][45]
Details in the story hint that a complex and multilayered editing process has been at work: the Exodus cities of Pithom and Rameses, for example, were not inhabited during most of the New Kingdom period, and the forty years of wilderness wanderings are also full of inconsistencies and anachronisms.[46] It is therefore best to treat the Exodus story not as the record of a single historical event but as a "powerful collective memory of the Egyptian occupation of Canaan and the enslavement of its population" during the 13th and 12th centuries (Ann Killebrew, 2005).[46] Extra-biblical accounts
The earliest non-Biblical account of the Exodus is in the writings of the Greek author Hecataeus of Abdera, who arrived in Egypt c.320 BCE; in his version the Egyptians blame a plague on foreigners and expel them from the country, whereupon Moses, their leader, takes them to Canaan.[47] The most famous is by the Egyptian historian Manetho (3rd century BCE), known from two quotations by the 1st century CE Jewish historian Josephus. In the first, Manetho describes the Hyksos, their lowly origins in Asia, their dominion over and expulsion from Egypt, and their subsequent foundation of the city of Jerusalem and its temple. Josephus (not Manetho) identifies the Hyksos with the Jews.[48] In the second story Manetho tells how 80,000 lepers and other "impure people", led by a priest named Osarseph, join forces with the former Hyksos, now living in Jerusalem, to take over Egypt. They wreak havoc until eventually the pharaoh and his son chase them out to the borders of Syria, where Osarseph gives the lepers a law-code and changes his name to Moses.[49] (The identification of Osarseph with Moses in the second account may be a later addition).[49][50]
This page was last modified on 24 February 2015
=========================================
Exodus of Israel from Egypt - REVEALED - Hard Evidence in Red Sea
DOWN WIND - Wind Farm documentary - FULL DOC in HD
2015/02/18 に公開
Down Wind is the explosive documentary that examines Ontario's controversial rush into wind farm development. Produced by Surge Media, Down Wind exposes how this Canadian provinces' green energy dream turned into a nightmare for rural residents forced to live among the towering 50 storey turbines. We hear searing, personal stories of people experiencing mysterious health problems, insomnia, depression, even thoughts of suicide; their lives turned upside down by the constant noise and vibrations given off by the massive wind turbines. The documentary also reveals the staggering economic costs of these wind farms to taxpayers with huge subsidies going to big wind corporations. And how inside connections have made some government cronies wealthy, while rural communities suffer. The film aired on Canada's Sun News Network. Media write up here: http://www.torontosun.com/2014/05/31/.... For more information contact: jeff.wigle@surgemedia.ca.
Comments
Here we report a unique cellular reprogramming phenomenon, called stimulus-triggered acquisition of pluripotency (STAP), which requires neither nuclear transfer nor the introduction of transcription factors. In STAP, strong external stimuli such as a transient low-pH stressor reprogrammed mammalian somatic cells, resulting in the generation of pluripotent cells. Through real-time imaging of STAP cells derived from purified lymphocytes, as well as gene rearrangement analysis, we found that committed somatic cells give rise to STAP cells by reprogramming rather than selection. STAP cells showed a substantial decrease in DNA methylation in the regulatory regions of pluripotency marker genes. Blastocyst injection showed that STAP cells efficiently contribute to chimaeric embryos and to offspring via germline transmission. We also demonstrate the derivation of robustly expandable pluripotent cell lines from STAP cells. Thus, our findings indicate that epigenetic fate determination of mammalian cells can be markedly converted in a context-dependent manner by strong environmental cues.
Figures
Figure 1: Stimulus-triggered conversion of lymphocytes into Oct4-GFP+ cells.
a, Schematic of low-pH treatment. b, Oct4-GFP+ cell clusters appeared in culture of low-pH-treated CD45+ cells (middle; high magnification, right) on day 7 (d7) but not in culture of control CD45+ cells (left). Top: bright-field view; bottom, GFP signals. Scale bar, 100μm. c, FACS analysis. The x axis shows CD45 epifluorescence level; y axis shows Oct4-GFP level. Non-treated, cultured in the same medium but not treated with low pH. d, GFP+ (green) and GFP− (yellow) cell populations (average cell numbers per visual field; ×10 objective lens). n = 25; error bars show average±s.d. e, Snapshots of live imaging of culture of low-pH-treated CD45+ cells (Oct4-gfp). Arrows indicate cells that started expressing Oct4-GFP. Scale bar, 50μm. f, Cell size reduction in low-pH-treated CD45+ cells on day1 before turning on Oct4-GFP without cell division on day 2. In this live imaging, cells were plated at a half density for easier viewing of individual cells. Scale bar, 10μm. g, Electron microscope analysis. Scale bar, 1μm. h, Forward scattering analysis of Oct4-GFP−CD45+ cells (red) and Oct4-GFP+CD45− cells (green) on day 7. Blue line, ES cells. i, Genomic PCR analysis of (D)J recombination at the Tcrb gene. GL is the size of the non-rearranged germline type, whereas the smaller ladders correspond to the alternative rearrangements of J exons. Negative controls, lanes 1, 2; positive controls, lane 3; FACS-sorted Oct4-GFP+ cells (two independent preparations on day 7), lanes 4, 5.
a, Immunostaining for pluripotent cell markers (red) in day 7 Oct4-GFP+ (green) clusters. DAPI, white. Scale bar, 50μm. b, qPCR analysis of pluripotency marker genes. From left to right, mouse ES cells; parental CD45+ cells; low-pH-induced Oct4-GFP+ cells on day 3; low-pH-induced Oct4-GFP+ cells on day 7. n = 3; error bars show average±s.d. c, DNA methylation study by bisulphite sequencing. Filled and open circles indicate methylated and non-methlylated CpG, respectively. d, Immunostaining analysis of in vitro differentiation capacity of day 7 Oct4-GFP+ cells. Ectoderm: the neural markers Sox1/Tuj1 (100%, n = 8) and N-cadherin (100%, n = 5). Mesoderm: smooth muscle actin (50%, n = 6) and brachyury (40%, n = 5). Endoderm: Sox17/E-cadherin (67%, n = 6) and Foxa2/Pdgfrα (67%, n = 6). Scale bar, 50μm. e, Teratoma formation assay of day 7 clusters of Oct4-GFP+ cells. Haematoxylin and eosin staining showed keratinized epidermis (ectoderm), skeletal muscle (mesoderm) and intestinal villi (endoderm), whereas immunostaining showed expression of Tuj1 (neurons), smooth muscle actin and α-fetoprotein. Scale bar, 100μm. f–i, Dissociation culture of ES cells and STAP cells (additional 7 days from day 7; f, g) on gelatin-coated dishes. Top, bright-field; bottom, alkaline phosphatase (AP) staining. Partially dissociated STAP cells slowly generated small colonies (i), whereas dissociated STAP cells did not, even in the presence of the ROCK inhibitor (g, h), which allows dissociation culture of EpiSCs29.
In the canalization view of Waddington’s epigenetic landscape, fates of somatic cells are progressively determined as cellular differentiation proceeds, like going downhill. It is generally believed that reversal of differentiated status requires artificial physical or genetic manipulation of nuclear function such as nuclear transfer1, 2 or the introduction of multiple transcription factors3. Here we investigated the question of whether somatic cells can undergo nuclear reprogramming simply in response to external triggers without direct nuclear manipulation. This type of situation is known to occur in plants—drastic environmental changes can convert mature somatic cells (for example, dissociated carrot cells) into immature blastema cells, from which a whole plant structure, including stalks and roots, develops in the presence of auxins4. A challenging question is whether animal somatic cells have a similar potential that emerges under special conditions. Over the past decade, the presence of pluripotent cells (or closely relevant cell types) in adult tissues has been a matter of debate, for which conflicting conclusions have been reported by various groups5, 6, 7, 8, 9, 10, 11. However, no study so far has proven that such pluripotent cells can arise from differentiated somatic cells.
Haematopoietic cells positive for CD45 (leukocyte common antigen) are typical lineage-committed somatic cells that never express pluripotency-related markers such as Oct4 unless they are reprogrammed12, 13. We therefore addressed the question of whether splenic CD45+ cells could acquire pluripotency by drastic changes in their external environment such as those caused by simple chemical perturbations.
CD45+ cells were sorted by fluorescence-activated cell sorting (FACS) from the lymphocyte fraction of postnatal spleens (1-week old) of C57BL/6 mice carrying an Oct4-gfp transgene14, and were exposed to various types of strong, transient, physical and chemical stimuli (described below). We examined these cells for activation of the Oct4 promoter after culture for several days in suspension using DMEM/F12 medium supplemented with leukaemia inhibitory factor (LIF) and B27 (hereafter called LIF+B27 medium). Among the various perturbations, we were particularly interested in low-pH perturbations for two reasons. First, as shown below, low-pH treatment turned out to be most effective for the induction of Oct4. Second, classical experimental embryology has shown that a transient low-pH treatment under ‘sublethal’ conditions can alter the differentiation status of tissues. Spontaneous neural conversion from salamander animal caps by soaking the tissues in citrate-based acidic medium below pH6.0 has been demonstrated previously15, 16, 17.
Without exposure to the stimuli, none of the cells sorted with CD45 expressed Oct4-GFP regardless of the culture period in LIF+B27 medium. In contrast, a 30-min treatment with low-pH medium (25-min incubation followed by 5-min centrifugation; Fig. 1a; the most effective range was pH5.4–5.8; Extended Data Fig. 1a) caused the emergence of substantial numbers of spherical clusters that expressed Oct4-GFP in day-7 culture (Fig. 1b). Substantial numbers of GFP+ cells appeared in all cases performed with neonatal splenic cells (n = 30 experiments). The emergence of Oct4-GFP+ cells at the expense of CD45+ cells was also observed by flow cytometry (Fig. 1c, top, and Extended Data Fig. 1b, c). We next fractionated CD45+ cells into populations positive and negative for CD90 (T cells), CD19 (B cells) and CD34 (haematopoietic progenitors18), and subjected them to low-pH treatment. Cells of these fractions, including T and B cells, generated Oct4-GFP+ cells at an efficacy comparable to unfractionated CD45+ cells (25–50% of surviving cells on day 7), except for CD34+ haematopoietic progenitors19, which rarely produced Oct4-GFP+ cells (<2%; Extended Data Fig. 1d).
a, Schematic of low-pH treatment. b, Oct4-GFP+ cell clusters appeared in culture of low-pH-treated CD45+ cells (middle; high magnification, right) on day 7 (d7) but not in culture of control CD45+ cells (left). Top: bright-field view; bottom, GFP signals. Scale bar, 100μm. c, FACS analysis. The x axis shows CD45 epifluorescence level; y axis shows Oct4-GFP level. Non-treated, cultured in the same medium but not treated with low pH. d, GFP+ (green) and GFP− (yellow) cell populations (average cell numbers per visual field; ×10 objective lens). n = 25; error bars show average±s.d. e, Snapshots of live imaging of culture of low-pH-treated CD45+ cells (Oct4-gfp). Arrows indicate cells that started expressing Oct4-GFP. Scale bar, 50μm. f, Cell size reduction in low-pH-treated CD45+ cells on day1 before turning on Oct4-GFP without cell division on day 2. In this live imaging, cells were plated at a half density for easier viewing of individual cells. Scale bar, 10μm. g, Electron microscope analysis. Scale bar, 1μm. h, Forward scattering analysis of Oct4-GFP−CD45+ cells (red) and Oct4-GFP+CD45− cells (green) on day 7. Blue line, ES cells. i, Genomic PCR analysis of (D)J recombination at the Tcrb gene. GL is the size of the non-rearranged germline type, whereas the smaller ladders correspond to the alternative rearrangements of J exons. Negative controls, lanes 1, 2; positive controls, lane 3; FACS-sorted Oct4-GFP+ cells (two independent preparations on day 7), lanes 4, 5.
Among maintenance media for pluripotent cells20, the appearance of Oct4-GFP+ cells was most efficient in LIF+B27 medium, and did not occur in mouse epiblast-derived stem-cell (EpiSC) medium21, 22 (Extended Data Fig. 1e). The presence or absence of LIF during days 0–2 did not substantially affect the frequency of Oct4-GFP+ cell generation on day 7 (Extended Data Fig. 1f), whereas the addition of LIF during days 4–7 was not sufficient, indicating that LIF dependency started during days 2–4.
Most of the surviving cells on day 1 were still CD45+ and Oct4-GFP−. On day3, the total cell numbers were reduced to between one-third to one-half of the day 0 population (Fig. 1d; see Extended Data Fig. 1g, h for apoptosis analysis), and a substantial number of total surviving cells became Oct4-GFP+ (Fig. 1d), albeit with relatively weak signal intensity. On day 7, a significant number of Oct4-GFP+CD45− cells (one-half to two-thirds of total surviving cells) constituted a distinct population from the Oct4-GFP−CD45− cells (Fig. 1c, top, day 7, and Fig. 1d). No obvious generation of Oct4-GFP+CD45− populations was seen in non-treated CD45+ cells cultured similarly but without low-pH treatment (Fig. 1c, bottom).
Low-pH-treated CD45+ cells, but not untreated cells, gradually turned on GFP signals over the first few days (Fig. 1e, Supplementary Videos 1 and 2 and Extended Data Fig. 2a), whereas CD45 immunoreactivity became gradually reduced in the cells that demonstrated Oct4-GFP expression (Fig. 1f and Extended Data Fig. 2b). By day 5, the Oct4-GFP+ cells attached together and formed clusters by accretion. These GFP+ clusters (but not GFP− cells) were quite mobile and often showed cell processes on moving (Supplementary Video 1).
The Oct4-GFP+ cells demonstrated a characteristic small cell size with little cytoplasm and also showed a distinct fine structure of the nucleus compared with that of parental CD45+ lymphocytes (Fig. 1g). The Oct4-GFP+ cells on day 7 were smaller than non-treated CD45+ cells (Fig. 1g, h and Extended Data Fig. 2c) and embryonic stem (ES) cells (Fig. 1h), both of which are generally considered to be small in size. The diameter of low-pH-treated CD45+ cells became reduced during the first 2days, even before they started Oct4-GFP expression (Fig. 1f), whereas the onset of GFP expression was not accompanied by cell divisions. Consistent with this, no substantial 5-ethynyl-2′-deoxyuridine (EdU) uptake was observed in the Oct4-GFP+ cells after the stressor (Extended Data Fig. 2d).
The lack of substantial proliferation argues against the possibility that CD45− cells, contaminating as a very minor population in the FACS-sorted CD45+ cells, quickly grew and formed a substantial Oct4-GFP+ population over the first few days after the low-pH treatment. In addition, genomic rearrangements of Tcrb (T-cell receptor gene) were observed in Oct4-GFP+ cells derived from FACS-purified CD45+ cells and CD90+CD45+ T cells (Fig. 1i, lanes 4, 5, and Extended Data Fig. 2e–g), indicating at least some contribution from lineage-committed T cells. Thus, Oct4-GFP+ cells were generated de novo from low-pH-treated CD45+ haematopoietic cells by reprogramming, rather than by simple selection of stress-enduring cells23.
On day 7, the Oct4-GFP+ spheres expressed pluripotency-related marker proteins22 (Oct4, SSEA1, Nanog and E-cadherin; Fig. 2a) and marker genes (Oct4, Nanog, Sox2, Ecat1 (also called Khdc3), Esg1 (Dppa5a), Dax1 (Nrob1) and Rex1 (Zfp42); Fig. 2b and Extended Data Fig. 3a) in a manner comparable to those seen in ES cells24. Moderate levels of expression of these pluripotency marker genes were observed on day 3 (Fig. 2b and Extended Data Fig. 3b). Notably, the Oct4-GFP+ cells on day 3, but not on day 7, expressed early haematopoietic marker genes such as Flk1 (also called Kdr) and Tal1 (Extended Data Fig. 3c), indicating that Oct4-GFP+ cells on day 3, as judged by their expression pattern at the population level, were still in a dynamic process of conversion.
a, Immunostaining for pluripotent cell markers (red) in day 7 Oct4-GFP+ (green) clusters. DAPI, white. Scale bar, 50μm. b, qPCR analysis of pluripotency marker genes. From left to right, mouse ES cells; parental CD45+ cells; low-pH-induced Oct4-GFP+ cells on day 3; low-pH-induced Oct4-GFP+ cells on day 7. n = 3; error bars show average±s.d. c, DNA methylation study by bisulphite sequencing. Filled and open circles indicate methylated and non-methlylated CpG, respectively. d, Immunostaining analysis of in vitro differentiation capacity of day 7 Oct4-GFP+ cells. Ectoderm: the neural markers Sox1/Tuj1 (100%, n = 8) and N-cadherin (100%, n = 5). Mesoderm: smooth muscle actin (50%, n = 6) and brachyury (40%, n = 5). Endoderm: Sox17/E-cadherin (67%, n = 6) and Foxa2/Pdgfrα (67%, n = 6). Scale bar, 50μm. e, Teratoma formation assay of day 7 clusters of Oct4-GFP+ cells. Haematoxylin and eosin staining showed keratinized epidermis (ectoderm), skeletal muscle (mesoderm) and intestinal villi (endoderm), whereas immunostaining showed expression of Tuj1 (neurons), smooth muscle actin and α-fetoprotein. Scale bar, 100μm. f–i, Dissociation culture of ES cells and STAP cells (additional 7 days from day 7; f, g) on gelatin-coated dishes. Top, bright-field; bottom, alkaline phosphatase (AP) staining. Partially dissociated STAP cells slowly generated small colonies (i), whereas dissociated STAP cells did not, even in the presence of the ROCK inhibitor (g, h), which allows dissociation culture of EpiSCs29.
On day 7, unlike CD45+ cells and like ES cells, low-pH-induced Oct4-GFP+ cells displayed extensive demethylation at the Oct4 and Nanog promoter areas (Fig. 2c), indicating that these cells underwent a substantial reprogramming of epigenetic status in these key genes for pluripotency. In vitro differentiation assays25, 26, 27 demonstrated that low-pH-induced Oct4-GFP+ cells gave rise to three-germ-layer derivatives (Fig. 2d) as well as visceral endoderm-like epithelium (Extended Data Fig. 3d). When grafted into mice, low-pH-induced Oct4-GFP+ cell clusters formed teratomas (40%, n = 20) (Fig. 2e and Extended Data Fig. 4a–c) but no teratocarcinomas that persistently contained Oct4-GFP+ cells (n = 50). Because some cellular variation was observed in the signal levels of Oct4-GFP within the clusters, we sorted GFP-strong cells (a major population) and GFP-dim cells (a minor population) by FACS on day 7 and separately injected them into mice. In this case, only GFP-strong cells formed teratomas (Extended Data Fig. 4d). In quantitative polymerase chain reaction (qPCR) analysis, the GFP-strong population expressed pluripotency marker genes but not early lineage-specific marker genes, whereas the GFP-dim cells showed substantial expression of some early lineage-specific marker genes (Flk1, Gata2, Gata4, Pax6 and Sox17; Extended Data Fig. 4e) but not Nanog and Rex1. These observations indicate that three-germ-layer derivatives were generated from the GFP-strong cells expressing pluripotency marker genes, rather than from GFP-dim cells that seem to contain partially reprogrammed cells.
Collectively, these findings show that the differentiation state of a committed somatic cell lineage can be converted into a state of pluripotency by strong stimuli given externally. Hereafter, we refer to the fate conversion from somatic cells into pluripotent cells by strong external stimuli such as low pH as ‘stimulus-triggered acquisition of pluripotency’ (STAP) and the resultant cells as STAP cells. Under their establishment conditions, these STAP cells were rarely proliferative (Extended Data Figs 2d and 5a, b). Comparative genomic hybridization array analysis of STAP cells indicated no major global changes in chromosome number (Extended Data Fig. 5c).
STAP cells, unlike mouse ES cells, showed a limited capacity for self-renewal in the LIF-containing medium and did not efficiently form colonies in dissociation culture (Fig. 2f, g), even in the presence of the ROCK inhibitor Y-27632, which suppresses dissociation-induced apoptosis28, 29 (Fig. 2h). Also, even under high-density culture conditions after partial dissociation (Fig. 2i), STAP cell numbers started to decline substantially after two passages. Furthermore, expression of the ES cell marker protein Esrrβ was low in STAP cells (Extended Data Fig. 5d, e). In general, female ES cells do not show X-chromosomal inactivation30 and contain no H3K27me3-dense foci (indicative of inactivated X chromosomes), unlike female CD45+ cells and EpiSCs. In contrast, H3K27me3-dense foci were found in ~40% of female STAP cells strongly positive for Oct4-GFP (Extended Data Fig. 5f, g).
STAP cells were also dissimilar to mouse EpiSCs, another category of pluripotent stem cell21, 22, 29, 31, and were positive for Klf4 and negative for the epithelial tight junction markers claudin 7 and ZO-1 (Extended Data Fig. 5d, e).
We next performed similar conversion experiments with somatic cells collected from brain, skin, muscle, fat, bone marrow, lung and liver tissues of 1-week-old Oct4-gfp mice. Although conversion efficacy varied, the low-pH-triggered generation of Oct4-GFP+ cells was observed in day 7 culture of all tissues examined (Fig. 3a and Extended Data Fig. 6a–c), including mesenchymal cells of adipose tissues (Fig. 3a–c) and neonatal cardiac cells that were negatively sorted for CD45 by FACS (Fig. 3d–g; see Extended Data Fig. 6d for suppression of cardiac genes such as Nkx2-5 and cardiac actin).
Figure 3: STAP cell conversion from a variety of cells by low-pH treatment.
a, Percentage of Oct4-GFP+ cells in day 7 culture of low-pH-treated cells from different origins (1×105 cells per ml×3ml). The number of surviving cells on day 7 compared to the plating cell number was 20–30%, except for lung, muscle and adipose cells, for which surviving cells were ~10% (n = 3, average±s.d.). b, Oct4-GFP+ cell clusters were induced by low-pH treatment from adipose-tissue-derived mesenchymal cells on day 7. Scale bar, 100μm. c, Expression of pluripotent cell markers in day 7 clusters of low-pH-treated adipose-tissue-derived mesenchymal cells. Scale bar, 50μm. d, Expression of pluripotency marker genes in STAP cells derived from various tissues. Gene expressions were normalized by Gapdh (n = 3, average±s.d.). Asterisk indicates adipose tissue-derived mesenchymal cells. e, Quantification of Oct4-GFP+ cells in culture of low-pH-treated neonatal cardiac muscle cells. ***P<0.001; Tukey’s test (n = 3). f, Generation of Oct4-GFP+ cell clusters (d7) from CD45− cardiac muscle cells. g, qPCR analysis of pluripotency marker genes in STAP cells from CD45− cardiac muscle cells.
We next performed a blastocyst injection assay with STAP cells that were generated from CD45+ cells of neonatal mice constitutively expressing GFP (this C57BL/6 line with cag-gfp transgenes is referred to hereafter as B6GFP). We injected STAP cell clusters en bloc that were manually cut into small pieces using a microknife (Fig. 4a). A high-to-moderate contribution of GFP-expressing cells was seen in the chimaeric embryos (Fig. 4b and Extended Data Fig. 7a). These chimaeric mice were born at a substantial rate and all developed normally (Fig. 4c and Extended Data Fig. 7b).
Figure 4: Chimaeric mouse generation from STAP cells.
a, Schematic of chimaeric mouse generation. b, E13.5 chimaera fetuses from 2N blastocytes injected with STAP cells (derived from B6GFP CD45+ cells carrying cag-gfp). c, Adult chimaeric mice generated by STAP-cell (B6GFP × 129/Sv; agouti) injection into blastocysts (ICR strain; albino). Asterisk indicates a highly contributed chimaeric mouse. d, Chimaera contribution analysis. Tissues from nine pups were analysed by FACS. e, Offspring of chimaeric mice derived from STAP cells. Asterisk indicates the same chimaeric mouse shown in c. f, E10.5 embryo generated in the tetraploid complementation assay with STAP cells (B6GFP×129/Sv).
CD45+ cell-derived STAP cells contributed to all tissues examined (Fig. 4d). Furthermore, offspring derived from STAP cells were born to the chimaeric mice (Fig. 4e and Extended Data Fig. 7c), demonstrating their germline transmission, which is a strict criterion for pluripotency as well as genetic and epigenetic normality32, 33. Furthermore, in a tetraploid (4N) complementation assay, which is considered to be the most rigorous test for developmental potency34, 35 (Fig. 4a, bottom), CD45+ cell-derived STAP cells (from F1 mice of B6GFP × 129/Sv or DBA/2) generated all-GFP+ embryos on embryonic day (E)10.5 (Fig. 4f, Extended Data Fig. 7d and Supplementary Video 3), demonstrating that STAP cells alone are sufficient to construct an entire embryonic structure. Thus, STAP cells have the developmental capacity to differentiate into all somatic-cell lineages as well as germ-cell lineages in vivo.
STAP cells have a limited self-renewal capacity under the conditions used for establishment (Fig. 2g and Extended Data Figs 2e and 5a). However, in the context of the embryonic environment, a small fragment of a STAP cell cluster could grow even into a whole embryo (Fig. 4f). With this in mind, we next examined whether STAP cells have the potential to generate expandable pluripotent cell lines in vitro under certain conditions.
STAP cells could not be efficiently maintained for additional passages in conventional LIF+FBS-containing medium or 2i medium20 (most STAP cells died in 2i medium within 7days; Extended Data Fig. 8a). Notably, an adrenocorticotropic hormone (ACTH)+LIF-containing medium (hereafter called ACTH medium) known to facilitate clonal expansion of ES cells36 supported outgrowth of STAP cell colonies. When cultured in this medium on a MEF feeder or gelatin, a portion of STAP cell clusters started to grow (Fig. 5a, bottom; such outgrowth was typically found in 10–20% of wells in single cluster culture using 96-well plates and in >75% when 12 clusters were plated per well). These growing colonies looked similar to those of mouse ES cells and expressed a high level of Oct4-GFP.
Figure 5: ES-cell-like stem cells can be derived from STAP cells.
a, Growth of STAP stem cells carrying Oct4-gfp. Scale bar, 50μm. b, Dissociation culture of STAP stem cells to form colonies. Scale bar, 100μm. c, Robust growth of STAP stem cells in maintenance culture. Similar results were obtained with eight independent lines. In contrast, parental STAP cells decreased in number quickly. d, Immunostaining of STAP stem cells for pluripotency markers (red). Scale bar, 50μm. e, qPCR analysis of pluripotency marker gene expression. f–h, In vitro differentiation assays into three-germ-layer derivatives. f, Ectoderm: Rx+/Pax6+ (retinal epithelium; 83%, n = 6). g, Mesoderm: troponin-T+ (cardiac muscle; 50%, n = 6). h, Endoderm: Sox17+/E-cadherin+ (endodermal progenitors; 67%, n = 6). Scale bar, 50μm. i, Teratoma formation assays. Formation of keratinized epidermis (ectoderm; left), cartilage (mesoderm; middle) and bronchial-like epithelium (endoderm; right) is shown. Scale bar, 100μm. j, Blastocyst injection assays. These pictures of live animals were taken serially (asterisk indicates the same chimaeric pup). k, l, Tetraploid complementation assay. ‘All-GFP+’ pups were born (k) and germline transmission was observed (l).
After culturing in ACTH medium for 7days, this growing population of cells, unlike parental STAP cells, could be passaged as single cells (Fig. 5a, bottom, and Fig. 5b), grow in 2i medium (Extended Data Fig. 8a) and expand exponentially, up to at least 120days of culture (Fig. 5c; no substantial chromosomal abnormality was seen; Extended Data Fig. 8b, c). Hereafter, we refer to the proliferative cells derived from STAP cells as STAP stem cells.
STAP stem cells expressed protein and RNA markers for pluripotent cells (Fig. 5d, e), showed low DNA methylation levels at the Oct4 and Nanog loci (Extended Data Fig. 8d), and had a nuclear fine structure similar to that of ES cells (Extended Data Fig. 8e; few electron-dense areas corresponding to heterochromatin). In differentiation culture25, 26, 27, STAP stem cells generated ectodermal, mesodermal and endodermal derivatives in vitro (Fig. 5f–h and Extended Data Fig. 8f, g), including beating cardiac muscles (Supplementary Video 4), and formed teratomas in vivo (Fig.5i and Extended Data Fig. 8h; no teratocarcinomas, n = 40). After blastocyst injection, STAP stem cells efficiently contributed to chimaeric mice (Fig. 5j), in which germline transmission was seen (Extended Data Fig. 8i). Even in tetraploid complementation assays, injected STAP stem cells could generate mice capable of growing to adults and producing offspring (Fig. 5k, l; in all eight independent lines, Extended Data Fig. 8j).
In addition to their expandability, we noticed at least two other differences between STAP stem cells and parental STAP cells. First, the expression of the ES cell marker protein Esrrβ, which was undetectable in STAP cells (Extended Data Fig. 5d, e), was clearly seen in STAP stem cells (Fig. 5e). Second, the presence of H3K27me3 foci, which was found in a substantial proportion of female STAP cells, was no longer observed in STAP stem cells (Extended Data Figs 5f and 8k). Thus, STAP cells have the potential to give rise to expandable cell lines that exhibit features similar to those of ES cells.
This study has revealed that somatic cells latently possess a surprising plasticity. This dynamic plasticity—the ability to become pluripotent cells—emerges when cells are transiently exposed to strong stimuli that they would not normally experience in their living environments.
Low-pH treatment was also used in the ‘autoneuralization’ experiment15, 16, 17 by Holtfreter in 1947, in which exposure to acidic medium caused tissue-autonomous neural conversion of salamander animal caps in vitro in the absence of Spemann’s organizer signals. Although the mechanism has remained elusive, Holtfreter hypothesized that the strong stimulus releases the animal cap cells from some intrinsic inhibitory mechanisms that suppress fate conversion or, in his words, they pass through ‘sublethal cytolysis’ (meaning stimulus-evoked lysis of the cell’s inhibitory state)15, 37. Although Holtfreter’s study and ours differ in the direction of fate conversion—orthograde differentiation and nuclear reprogramming, respectively—these phenomena may share some common aspects, particularly with regard to sublethal stimulus-evoked release from a static (conversion-resisting) state in the cell.
A remaining question is whether cellular reprogramming is initiated specifically by the low-pH treatment or also by some other types of sublethal stress such as physical damage, plasma membrane perforation, osmotic pressure shock, growth-factor deprivation, heat shock or high Ca2+ exposure. At least some of these stressors, particularly physical damage by rigorous trituration and membrane perforation by streptolysin O, induced the generation of Oct4-GFP+ cells from CD45+ cells (Extended Data Fig. 9a; see Methods). These findings raise the possibility that certain common regulatory modules, lying downstream of these distantly related sublethal stresses, act as a key for releasing somatic cells from the tightly locked epigenetic state of differentiation, leading to a global change in epigenetic regulation. In other words, unknown cellular functions, activated by sublethal stimuli, may set somatic cells free from their current commitment to recover the naive cell state.
Our present finding of an unexpectedly large capacity for radical reprogramming in committed somatic cells raises various important questions. For instance, why, and for what purpose, do somatic cells latently possess this self-driven ability for nuclear reprogramming, which emerges only after sublethal stimulation, and how, then, is this reprogramming mechanism normally suppressed? Furthermore, why isn’t teratoma (or pluripotent cell mass) formation normally seen in in vivo tissues that may receive strong environmental stress? In our preliminary study, experimental reflux oesophagitis locally induced moderate expression of Oct4-GFP but not endogenous Nanog in the mouse oesophageal mucosa (Extended Data Fig. 9b). Therefore, an intriguing hypothesis for future research is that the progression from initial Oct4 activation to further reprogramming is suppressed by certain inhibitory mechanisms in vivo.
The question of why and how this self-driven reprogramming is directed towards the pluripotent state is fundamentally important, given that STAP reprogramming takes a remarkably short period, only a few days for substantial expression of pluripotency marker genes, unlike transgene- or chemical-induced iPS cell conversion38. Thus, our results cast new light on the biological meaning of diverse cellular states in multicellular organisms.
Research involving animals complied with protocols approved by the Harvard Medical School/Brigham and Women’s Hospital Committee on Animal Care, and the Institutional Committee of Laboratory Animal Experimentation of the RIKEN Center for Developmental Biology.
Tissue collection and low-pH treatment
To isolate CD45+ haematopoietic cells, spleens were excised from 1-week-old Oct4-gfp mice (unless specified otherwise), minced by scissors and mechanically dissociated with pasture pipettes. Dissociated spleen cells were suspended with PBS and strained through a cell strainer (BD Biosciences). After centrifuge at 1,000r.p.m. for 5min, collected cells were re-suspended in DMEM medium and added to the same volume of lympholyte (Cedarlane), then centrifuged at 1,000g for 20min. The lymphocyte layer was taken out and stained with CD45 antibody (ab25603, Abcam). CD45-positive cells were sorted by FACS Aria (BD Biosciences). After cell sorting, 1× 106 CD45-positive cells were treated with 500μl of low-pH HBSS solution (titrated to pH5.7 by HCl) for 25min at 37°C, and then centrifuged at 1,000r.p.m. at room temperature for 5min. After the supernatant (low-pH solution) was removed, precipitated cells were re-suspended and plated onto non-adhesive culture plates (typically, 1×105 cellsml−1) in DMEM/F12 medium supplemented with 1,000U LIF (Sigma) and 2% B27 (Invitrogen). Cell cluster formation was more sensitive to the plating cell density than the percentage of Oct4-GFP+ cells. The number of surviving cells was sensitive to the age of donor mice and was low under the treatment conditions above when adult spleens were used. The addition of LIF during days 2–7 was essential for generating Oct4-GFP+ STAP cell clusters on day 7, as shown in Extended Data Fig. 1f. Even in the absence of LIF, Oct4-GFP+ cells (most of them were dim in signal) appeared transiently during days 2–5 in culture of low-pH-treated CD45+ cells, but subsequently disappeared, indicating that there is a LIF-independent early phase, whereas the subsequent phase is LIF-dependent.
Chimaeric mouse generation and analyses
For production of diploid and tetraploid chimaeras with STAP cells, diploid embryos were obtained from ICR strain females. Tetraploid embryos were produced by electrofusion of 2-cell embryos. Because trypsin treatment of donor samples turned out to cause low chimaerism, STAP spherical colonies were cut into small pieces using a microknife under the microscope, and small clusters of STAP cells were then injected into day-4.5 blastocysts by a large pipette. The next day, the chimaeric blastocysts were transferred into day-2.5 pseudopregnant females. For experiments using STAP cells from CD45+ cells without the Oct4-gfp reporter, STAP cell clusters were identified by their characteristic cluster morphology (they are made of very small cells with no strong compaction in the aggregate). When the STAP conversion conditions (low pH) were applied to CD45+ lymphocytes, most day-7 clusters that were large and contained more than a few dozen small cells were positive for Oct4 (although the expression level varied). Therefore, we used only well-formed characteristic clusters (large ones) for this type of study and cut them by microknife to prepare donor cell clusters in a proper size for glass needle injection. For an estimate of the contribution of these injected cells, we used STAP cells that were generated from CD45+ cells of mice constitutively expressing GFP (C57BL/6 line with cag-gfp transgenes; F1 of C57BL/6 and 129/Sv or DBA/2 was used from the viewpoint of heterosis).
Because the number of CD45+ cells from a neonatal spleen was small, we mixed spleen cells from male and female mice for STAP cell conversion. To make germline transmission more efficient, we intercrossed chimaeras in some experiments.
For the production of diploid and tetraploid chimaeras with STAP stem cells, diploid embryos were obtained from ICR strain females. Tetraploid embryos were produced by electrofusion of 2-cell embryos. STAP stem cells were dissociated into single cells and injected into day-4.5 blastocysts. In the chimaera studies with both STAP cells and STAP stem cells, we did not find tumorigenetic tendencies in their chimaeras or their offspring (up to 18months).
In vivo differentiation assay
1×107 STAP cells were seeded onto a sheet composed of a non-woven mesh of polyglycolic acid fibres (3× 3×1mm; 200μm in pore diameter), cultured for 24h in DMEM + 10% FBS, and implanted subcutaneously into the dorsal flanks of 4-week-old mice. In this experiment, to better support tumour formation from slow growing STAP cells by keeping cells in a locally dense manner, we implanted STAP cells with artificial scaffold made of polyglycolic acid fibres. Given the artificial nature of the material, we used NOD/SCID mice as hosts, to avoid possible enhancement of post-graft inflammation caused by this scaffold even in syngenic mice. STAP stem cells were dissociated into single cells and cell suspension containing 1×107 cells was injected into the testis. Six weeks later, the implants were analysed using histochemical techniques. The implants were fixed with 10% formaldehyde, embedded in paraffin, and routinely processed into 4-µm-thick sections. Sections were stained with haematoxylin and eosin. Endoderm tissues were identified with expression of anti-α-fetoprotein (mouse monoclonal antibody; MAB1368, R&D Systems). Ectodermal tissues were identified with expression of anti-βIII tubulin (mouse monoclonal antibody; G7121, Promega). Mesodermal tissues were identified with expression of anti-α-smooth muscle actin (rabbit polyclonal; DAKO). In negative controls, the primary antibody was replaced with IgG-negative controls of the same isotype to ensure specificity.
STAP by exposure to other external stimuli
To give a mechanical stress to mature cells, a pasture pipette was heated and then stretched to create thin capillaries with the lumens approximately 50μm in diameter, and then broken into appropriate lengths. Mature somatic cells were then repeatedly triturated through these pipettes for 20min, and then cultured for 7days. To provide a heat shock, cells were heated at 42°C for 20min and cultured for 7days. A nutrition-deprivation stress was provided to mature cells, by culturing the cells in basal culture medium for 3weeks. High Ca2+ concentration stress was provided to mature cells by culturing cells in medium containing 2mM CaCl2 for 7days. To give a strong stress by creating pores in cell membranes, cells were treated with 230ngml−1 streptolysine O (SLO) (S5265, Sigma) for 2h, then cultured for 7days. After each treatment, the ratio of Oct4-GFP-positive cells was analysed by FACS.
Bisulphite sequencing
GFP-positive cells in STAP clusters were collected by FACS Aria. Genomic DNA was extracted from STAP cells and analysed. Bisulphite treatment of DNA was performed using the CpGenome DNA modification kit (Chemicon, http://www.chemicon.com), following the manufacturer’s instructions. The resulting modified DNA was amplified by nested PCR using two forward (F) primers and one reverse (R) primer: Oct4 (F1, 5′-GTTGTTTTGTTTTGGTTTTGGATAT-3′; F2, 5′-ATGGGTTGAAATATTGGGTTTATTTA-3′; R, 5′-CCACCCTCTAACCTTAACCTCTAAC-3′). And Nanog (F1, 5′-GAGGATGTTTTTTAAGTTTTTTTT-3′; F2, 5′-AATGTTTATGGTGGATTTTGTAGGT-3′; R, 5′-CCCACACTCATATCAATATAATAAC-3′). PCR was done using TaKaKa Ex Taq Hot Start Version (RR030A). DNA sequencing was performed using a M13 primer at the Genome Resource and Analysis Unit, RIKEN CDB.
Immunohistochemistry
Cultured cells were fixed with 4% paraformaldehyde and permeabilized with 0.1% Triton X-100/PBS before blocking with 1% BSA solution. Cells were incubated with the following primary antibodies: anti-Oct4 (Santa Cruz Biotechnology; C-10), anti-Nanog (eBioscience; MLC-51), anti-SSEA-1 (Millipore; MC480), anti-E-cadherin (Abcam), anti-ZO-1 (Santa Cruz Biotechnology; c1607), anti-claudin7 (Abcam), anti-Klf4 (R&D Systems), anti-Esrrβ (R&D Systems), anti-H3K27me3 (Millipore), anti-BrdU (BD Bioscience) and anti-Ki67 (BD Pharmingen). After overnight incubation, cells were incubated with secondary antibodies: goat anti-mouse or -rabbit coupled to Alexa-488 or -594 (Invitrogen). Cell nuclei were visualized with DAPI (Sigma). Slides were mounted with a SlowFade Gold antifade reagent (Invitrogen).
Fluorescence-activated cell sorting and flow cytometry
Cells were prepared according to standard protocols and suspended in 0.1% BSA/PBS on ice before FACS. Propidium iodide (BD Biosciences) was used to exclude dead cells. In negative controls, the primary antibody was replaced with IgG-negative controls of the same isotype to ensure specificity. Cells were sorted on a BD FACSAria SORP and analysed on a BD LSRII with BD FACS Diva Software (BD Biosciences). For haematopoietic fraction sorting, antibodies against T-cell marker (anti-CD90; eBioscience), B-cell marker (anti-CD19; Abcam) and haematopoietic progenitor marker (anti-CD34; Abcam) were used.
RNA preparation and RT–PCR analysis
RNA was isolated with the RNeasy Micro kit (Qiagen). Reverse transcription was performed with the SuperScript III first strand synthesis kit (Invitrogen). Power SYBR Green Mix (Roche Diagnostics) was used for amplification, and samples were run on a Lightcycler-II Instrument (Roche Diagnostics). The primer sets for each gene are listed in Supplementary Table 1.
In vitro differentiation assays
For mesodermal differentiation assay, STAP cells were collected at 7days, and Oct4-GFP-positive cells were collected by cell sorter and subjected to culture in DMEM supplemented with 20% FBS. Medium was exchanged every 3days. After 7–14days, muscle cells were stained with an anti-α-smooth muscle actin antibody (DAKO).
For neural lineage differentiation assay, STAP cells were collected at 7days and subjected to SDIA or SFEBq culture. For SDIA culture, collected STAP cell clusters were plated on PA6 cell feeder as described previously26. For SFEBq culture, STAP cell clusters (one per well; non-cell-adhesive 96-well plate, PrimeSurface V-bottom, Sumitomo Bakelite) were plated and cultured in suspension as described previously36.
For endodermal differentiation, STAP cells were collected at 7days and subjected to suspension culture with inducers in 96-well plates27.
TCR-β chain gene rearrangement analysis
Genomic DNA was extracted from STAP cells and tail tips from chimaeric mice generated with STAP cells derived from CD45+ cells. PCR was performed with 50ng DNA using the following primers (Dβ2: 5′-GCACCTGTGGGGAAGAAACT-3′ and Jβ2.6: 5′-TGAGAGCTGTCTCCTACTATCGATT-3′) that amplify the regions of the (D)J recombination. The PCR products were subjected to gel electrophoresis in Tris-acetate-EDTA buffer with 1.6% agarose and visualized by staining with ethidium bromide. PCR bands from STAP cells were subjected to sequencing analysis and identified as rearranged genomic fragments of the (D)J recombination.
EdU uptake assay and apoptosis analysis
At various phases in STAP cell culture (days 0–2, 2–7, 7–14), EdU was added to the culture medium (final concentration: 10μM) and EdU uptake was analysed by FACS. This assay was performed according to the manufacturer’s protocol with the Click-iT EdU Flow cytometry assay kit (Invitrogen).
Apoptosis analysis was performed with flow cytometry using Annexin-V (Biovision) and propidium iodide. Annexin-V analysis by FACS on day 14 showed that most Oct4-GFP+ cells were positive for this apoptotic marker; indeed, the number of surviving cells declined thereafter.
Soft agar assay
Sorted STAP cells (Oct4-GFP-strong or -dim) and control mouse ES cells (1,000 cells per well of 96-well plate) were plated into soft ager medium (0.4% agarose) in LIF-B27 medium. After 7days of culture, cells were dissociated and their anchorage-independent growth was quantified by fluorescent measurement with the cytoselect 96-well cell transformation assay kit (Cell Biolabs) according to the manufacturer’s protocol.
Genomic DNA was extracted from STAP (male) and CD45-positive cells (male) by the Gene JET Genomic DNA purification kit (Thermo Scientific). Using CGH array (Agilent), the normality of chromosomes derived from STAP was compared with that of CD45-positive cells whose chromosomal normality was confirmed by a separate experiment. CGH array and data analysis were performed at TAKARA Bio.
Electron microscopy
For electron microscopic analysis, dissociated cells were fixed in 2.5% glutaraldehyde and 2% formaldehyde in 0.1M cacodylate buffer (pH7.2) and then processed for thin sectioning and transmission electron microscopy.
Live cell imaging
All live-cell imaging was performed with LCV110-CSUW1 (Olympus). For live-cell imaging of ‘in culture CD45 antibody staining’, CD45+ cells treated with low pH were plated in culture medium containing 20ngml−1 of fluorescent-labelled CD45 antibody (eBioscience)40.
RNA-sequencing and ChIP sequencing analyses
For RNA sequencing of cell lines, total RNA was extracted from cells by the RNasy mini kit (Qiagen). RNA-seq libraries were prepared from 1μg total RNAs following the protocol of the TruSeq RNA Sample Prep kit (Illumina) and subjected to the deep sequencing analysis with Illumina Hi-Seq1500. Cluster tree diagram of various cell types was obtained from hierarchical clustering of global expression profiles (log2 FPKM of all transcripts; FPKM, fragments per kilobase of transcript per million mapped reads). Complete linkage method applied to 1−r (r = Pearson’s correlation between profiles) was used for generating the tree and 1,000 cycles of bootstrap resampling were carried out to obtain statistical confidence score in per cent units (also called AU P values).
ChIP-seq libraries were prepared from 20ng input DNAs, 1ng H3K4me3 ChIP DNAs, or 5ng H3K27me3 ChIP DNAs using the KAPA Library Preparation kit (KAPA Biosystems). TruSeq adaptors were prepared in-house by annealing a TruSeq universal oligonucleotide and each of index oligonucleotides (5′-AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACACGACGCTCTTCCGATCT-3′, and 5′-GATCGGAAGAGCACACGTCTGAACTCCAGTCACXXXXXXATCTCGTATGCCGTCTTCTGCTTG-3′; where X represents index sequences).
Chromatin immunoprecipitation was performed as follows. Cells were fixed in PBS(-) containing 1% formaldehyde for 10min at room temperature. Glycine was added to a final concentration of 0.25M to stop the fixation. After washing the cells twice in ice-cold PBS(-), cells were further washed in LB1 (50mM HEPES-KOH pH7.5, 140mM NaCl, 1mM EDTA, 10% glycerol, 0.5% NP-40, 0.25% Triton X-100) and LB2 (10mM Tris-HCl pH8.0, 200mM NaCl, 1mM EDTA, 0.5mM EGTA). Cells were then re-suspended in lysis buffer (50mM Tris-HCl pH8.0, 10mM EDTA, 1% SDS). Lysates were prepared by sonication using Covaris S220 in a mini tube at duty cycle = 5%, PIP = 70, cycles per burst = 200, and the treatment time of 20min. Lysates from 2×106 cells were diluted in ChIP dilution buffer (16.7mM Tris-HCl pH8.0, 167mM NaCl, 1.2mM EDTA, 1.1% Triton X-100, 0.01% SDS). ChIP was performed using sheep anti-mouse IgG beads (Invitrogen) or protein A beads (Invitrogen) coupled with anti-histone H3K4me3 antibody (Wako, catalogue no. 307-34813) or anti-histone H3K27me3 antibody (CST, catalogue no. 9733), respectively. After 4–6h of incubation in a rotator at 4°C, beads were washed five times in low-salt wash buffer (20mM Tris HCl pH8.0, 150mM NaCl, 2mM EDTA, 1% Triton X-100, 0.1% SDS), and three times in high-salt wash buffer (20mM Tris-HCl pH8.0, 500mM NaCl, 2mM EDTA, 1% Triton X-100, 0.1% SDS). Target chromatin was eluted off the beads in elution buffer (10mM Tris-HCl pH8.0, 300mM NaCl, 5mM EDTA, 1% SDS) at room temperature for 20min. Crosslink was reversed at 65°C, and then samples were treated with RNaseA and proteinase K. The prepared DNA samples were purified by phenol-chloroform extraction followed by ethanol precipitation and dissolved in TE buffer.
STAP stem-cell conversion culture
For establishment of STAP stem-cell lines, STAP cell clusters were transferred to ACTH-containing medium36 on MEF feeder cells (several clusters, up to a dozen clusters, per well of 96-well plates). Four to seven days later, the cells were subjected to the first passage using a conventional trypsin method, and suspended cells were plated in ES maintain medium containing 20% FBS. Subsequent passaging was performed at a split ratio of 1:10 every second day before they reached subconfluency. We tested the following three different genetic backgrounds of mice for STAP stem-cell establishment from STAP cell clusters, and observed reproducible data of establishment: C57BL/6 carrying Oct4-gfp (29 of 29), 129/Sv carrying Rosa26-gfp (2 of 2) and 129/Sv×C57BL/6 carrying cag-gfp (12 of 16). STAP stem cells with all these genetic backgrounds showed chimaera-forming activity.
For clonal analysis of STAP stem cells, single STAP stem cells were manually picked by a thin-glass pipette, and plated into 96-well plates at one cell per well. The clonal colonies were cultured in ES medium containing 20% FBS, and expanded for subsequent experiments.
Karyotype analysis
Karyotype analysis was performed by Multicolor FISH analysis (M-FISH). Subconfluent STAP stem cells were arrested in metaphase by colcemid (final concentration 0.270µgml−1) to the culture medium for 2.5h at 37°C in 5% CO2. Cells were washed with PBS, treated with trypsin and EDTA (EDTA), re-suspended into cell medium and centrifuged for 5min at 1,200r.p.m. To the cell pellet in 3ml of PBS, 7ml of a pre-warmed hypotonic 0.0375M KC1 solution was added. Cells were incubated for 20min at 37°C. Cells were centrifuged for 5min at 1,200r.p.m. and the pellet was re-suspended in 3–5ml of 0.0375M KC1 solution. The cells were fixed with methanol/acetic acid (3:1; vol/vol) by gently pipetting. Fixation was performed four times before spreading the cells on glass slides. For the FISH procedure, mouse chromosome-specific painting probes were combinatorially labelled using seven different fluorochromes and hybridized as previously described41. For each cell line, 9–15 metaphase spreads were acquired by using a Leica DM RXA RF8 epifluorescence microscope (Leica Mikrosysteme GmbH) equipped with a Sensys CCD camera (Photometrics). Camera and microscope were controlled by the Leica Q-FISH software (Leica Microsystems). Metaphase spreads were processed on the basis of the Leica MCK software and presented as multicolour karyograms.
Q-band analysis was performed at Chromocentre (Japan). After quinacrin staining, 20 cells from each sample were randomly selected and the normality of chromosomes was analysed. Five different independent lines of STAP stem cells showed no chromosomal abnormalities in Q-band analysis after >10 passages.
Gurdon, J. B.The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles. J. Embryol. Exp. Morphol.10, 622–640 (1962)
Wakayama, T., Perry, A. C., Zuccotti, M., Johnson, K. R. & Yanagimachi, R.Full-term development of mice from enucleated oocytes injected with cumulus cell nuclei. Nature394, 369–374 (1998)
Takahashi, K. & Yamanaka, S.Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell126, 663–676 (2006)
D’Ippolito, G.et al.Marrow-isolated adult multilineage inducible (MIAMI) cells, a unique population of postnatal young and old human cells with extensive expansion and differentiation potential. J. Cell Sci.117, 2971–2981 (2004)
Kucia, M.et al.A population of very small embryonic-like (VSEL) CXCR4+SSEA-1+Oct-4+ stem cells identified in adult bone marrow. Leukemia20, 857–869 (2006)
Ohbo, K.et al.Identification and characterization of stem cells in prepubertal spermatogenesis in mice small star, filled. Dev. Biol.258, 209–225 (2003)
Gratama, J. W., Sutherland, D. R. & Keeney, M.Flow cytometric enumeration and immunophenotyping of hematopoietic stem and progenitor cells. Semin. Hematol.38, 139–147 (2001)
Ohgushi, M.et al.Molecular pathway and cell state responsible for dissociation-induced apoptosis in human embryonic stem cells. Cell Stem Cell7, 225–239 (2010)
Murakami, K., Araki, K., Ohtsuka, S., Wakayama, T. & Niwa, H.Choice of random rather than imprinted X inactivation in female embryonic stem cell-derived extra-embryonic cells. Development138, 197–202 (2011)
Nagy, A., Rossant, J., Nagy, R., Abramow-Newerly, W. & Roder, J. C.Derivation of completely cell culture-derived mice from early-passage embryonic stem cells. Proc. Natl Acad. Sci. USA90, 8424–8428 (1993)
Eakin, G. S., Hadjantonakis, A. K., Papaioannou, V. E. & Behringer, R. R.Developmental potential and behavior of tetraploid cells in the mouse embryo. Dev. Biol.288, 150–159 (2005)
Jentsch, I., Geigl, J., Klein, C. A. & Speicher, M. R.Seven-fluorochrome mouse M-FISH for high-resolution analysis of interchromosomal rearrangements. Cytogenet. Genome Res.103, 84–88 (2003)
We thank S. Nishikawa for discussion and J. D. Ross, N. Takata, M. Eiraku, M. Ohgushi, S. Itoh, S. Yonemura, S. Ohtsuka and K. Kakiguchi for help with experiments and analyses. We thank A. Penvose and K. Westerman for comments on the manuscript. H.O. is grateful to T. Okano, S. Tsuneda and K. Kuroda for support and encouragement. Financial support for this research was provided by Intramural RIKEN Research Budget (H.O., T.W. and Y.S.), a Scientific Research in Priority Areas (20062015) to T.W., the Network Project for Realization of Regenerative Medicine to Y.S., and Department of Anesthesiology, Perioperative and Pain Medicine at Brigham and Women’s Hospital to C.A.V.
Laboratory for Tissue Engineering and Regenerative Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
Haruko Obokata,
Koji Kojima,
Martin P. Vacanti &
Charles A. Vacanti
Laboratory for Cellular Reprogramming, RIKEN Center for Developmental biology, Kobe 650-0047, Japan
Haruko Obokata
Laboratory for Genomic Reprogramming, RIKEN Center for Developmental biology, Kobe 650-0047, Japan
Haruko Obokata &
Teruhiko Wakayama
Laboratory for Organogenesis and Neurogenesis, RIKEN Center for Developmental biology, Kobe 650-0047, Japan
Yoshiki Sasai
Department of Pathology, Irwin Army Community Hospital, Fort Riley, Kansas 66442, USA
Martin P. Vacanti
Laboratory for Pluripotent Stem Cell Studies, RIKEN Center for Developmental biology, Kobe 650-0047, Japan
Hitoshi Niwa
Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Tokyo 162-8666, Japan
Masayuki Yamato
Present address: Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan.
Teruhiko Wakayama
Contributions
H.O. and Y.S. wrote the manuscript. H.O., T.W. and Y.S. performed experiments, and K.K. assisted with H.O.’s transplantation experiments. H.O., T.W., Y.S., H.N. and C.A.V. designed the project. M.P.V. and M.Y. helped with the design and evaluation of the project.
Competing financial interests
The authors declare no competing financial interests.
1.Extended Data Figure 1: Conversion of haematopoietic cells into Oct4-GFP+ cells by a low-pH exposure. (292 KB) a, Optimization of pH conditions for Oct4-GFP induction. Five days after CD45-positive cells were exposed to acidic solution treatment at different pH, Oct4-GFP expression was analysed by FACS (n = 3, average±s.d.). b, Gating strategy for Oct4-GFP+ cell sorting. Top: representative results 7days after the stress treatment. Bottom: non-treated control. P3 populations were sorted and counted as Oct4-GFP+ cells for all experiments. c, Controls for FACS analysis. In Oct4-GFP+ cell analysis, the grey and white histograms indicate the negative control (non-stress-treated Oct4-gfp haematopoietic cells) and the positive control (Oct4-gfp ES cells), respectively. Also, the green histograms indicate non-treated cells (left) and stress-treated cells at day7 (right). In CD45+ cell analysis, the grey and white histograms indicate the negative (isotype) and positive controls, respectively. The red histograms indicate non-stress-treated cells (left) and stress-treated cells at day7 (right). d, Oct4-GFP+ cell generation from various subpopulations of CD45+ cells. Seven days after the stress treatment, Oct4-GFP expression was analysed by FACS (n = 3, average±s.d.). Among total CD45+ fraction and its subfractions of CD19+, CD90+, CD34+ and CD34− cells, the efficacy of CD34+ cells was significantly lower than the others. P<0.05 by the Newman–Keuls test and P<0.01 by one-way ANOVA. e, Comparison of culture conditions for low-pH-induced conversion. Stress-treated cells were cultured in various media. The number of Oct4-GFP-expressing clusters was counted at day 14 (n = 3, average±s.d.). ***P<0.001 (B27+LIF versus all other groups); Tukey’s test. In the case of 3i medium, although the clusters appeared at a moderate efficiency, they appeared late and grew slowly. ACTH, ACTH-containing ES medium; ES+LIF+FBS, 20% FBS+LIF-containing ES culture medium; B27, DMEM/F12 medium containing 2% B27; B27+LIF, DMEM/F12 medium containing 2% B27+LIF; EpiSC, EpiSC culture medium containing Fgf2+activin. f, Signalling factor dependency of STAP cell generation. Growth factors that are conventionally used for pluripotent cell culture such as LIF, activin, Bmp4 and Fgf2 were added to basal culture medium (B27-supplemented DMEM/F12) in different culture phases (days 0–7, 2–7 and 4–7), and Oct4-GFP expression was analysed by FACS at day 7 (n = 3, average±s.d.). g, h, Time course of apoptosis after the low-pH exposure. Stress-treated cells and non-stress-treated control cells were stained with CD45, annexin-V and propidium iodide at day 0 (immediately after stress treatment), day 3 and day 7. g, Blue bars, GFP+CD45−; orange bars, GFP−CD45+. Percentages in total cells included propidium-iodide-positive cells. h, Annexin-V-positive cells in these cell populations were analysed by FACS.
2.Extended Data Figure 2: Phenotypic change during STAP cell conversion. (231 KB) a, Oct4 protein expression in STAP cells was detected by immunostaining at day 2 (left) and day 7 (right). b, Live cell imaging of STAP conversion (grey, CD45 antibody; green, Oct4-GFP). See Methods for experimental details to monitor live CD45 immunostaining. c, Immunostaining of a parental CD45+ cell (left) and an Oct4-GFP+ cell (right). Scale bar, 10μm. d, EdU uptake assay (n = 3, average±s.d.). e, Schematic of Tcrb gene rearrangement. f, T-cell-derived STAP cells. Scale bar, 100μm. g, Genomic PCR analysis of (D)J recombination at the Tcrb gene of T-cell-derived STAP cells. G.L. is the size of the non-rearranged germline type, whereas the smaller ladders correspond to the alternative rearrangements of J exons (confirmed by sequencing). Negative controls (ES cells), positive controls (lymphocytes) and T-cell-derived STAP (two independent preparations on d7) are indicated.
3.Extended Data Figure 3: Gene expression analyses during STAP conversion and endoderm differentiation assay. (213 KB) a, Expression of pluripotency marker genes in STAP cells derived from T cells (n = 3, average±s.d.). b, Expression of pluripotency marker genes in STAP cells. In this experiment, Oct4-GFP+ cells seen in live cell imaging (Extended Data Fig. 2b) were analysed to confirm their conversion into STAP cells (n = 3, average±s.d.). c, Haematopoietic marker expression during STAP conversion from CD45+ cells (n = 3, average±s.d.). d, Formation of visceral endoderm-like surface epithelium in differentiating STAP cluster on day 2 (left) and day 8 (right). Scale bars, 50μm.
4.Extended Data Figure 4: Teratoma formation assay and characterization of Oct4-GFP-dim cells. (265 KB) a–c, Teratomas formed from STAP cell clusters included neuroepithelium (a), striated muscle (b) and pancreas (c; right, high-magnification view showing a typical acinar morphology and ductal structures). Scale bars, 100μm. d, Teratoma-forming ability of Oct4-GFP+ and Oct4-GFP-dim cells (isolated by FACS, top). Oct4-GFP+ cells, but not Oct4-GFP-dim cells, efficiently formed teratomas (table at the bottom). However, because STAP cells were dissociation-intolerant, the teratoma-forming efficiency of dissociated Oct4-GFP+ cells was lower than that of non-dissociated STAP cell clusters. e, Gene expression of Oct4-GFP+ and Oct4-GFP-dim cells (n = 3, the average±s.d.). Haematopoietic marker gene expression (left) and early lineage marker gene expression (right) are shown.
5.Extended Data Figure 5: In vitro characterization of STAP cells. (430 KB) a, Immunostaining for Ki67 and BrdU. STAP cell clusters (top) and ES cell colonies (bottom) are shown. For BrdU uptake, BrdU was added into each culture medium (10μM) for 12h until fixation. Scale bar, 100μm. b, Transformation assay by soft agar culture. Neither Oct4-GFP+ nor Oct4-GFP-dim cells showed colony formation in soft agar, whereas ES cells and STAP stem cells showed anchorage-independent growth in the same LIF-B27 medium. Scale bar, 100µm. Proliferated cells were lysed and the amount of DNA in each well was estimated by chemical luminescence (graph). n = 3 , average±s.d. c, No substantial change in chromosome number was seen with STAP cells in the CGH array. Genomic DNA derived from CD45+ cells (male) was used as reference DNA. The spikes (for example, those seen in the X chromosome) were nonspecific and also found in the data of these parental CD45+ cells when the manufacturer’s control DNA was used as a reference. d, qPCR analysis for pluripotency markers that highly express in ES cells, but not in EpiSCs. Average±s.d. e, Immunostaining of markers for mouse EpiSC and ES cells. Scale bar, 100 μm. f, g, H3K27me3+ foci in female cells, which are indicative of X-chromosomal inactivation. These foci were not observed in male cells. Scale bar, 10μm. In the case of female STAP cells, ~40% of cells retained H3K27me3+ foci (g). **P<0.001; Tukey’s test. n = 3, average±s.d. Although nuclear staining looked to be higher in STAP cells with H3K27me3+ foci (f), this appeared to be caused by some optical artefacts scattering from the strong focal signal. h, qPCR analysis for the tight junction markers Zo-1 and claudin 7, which were highly expressed in EpiSCs, but not in ES cells or STAP cells. **P<0.01; ns, not significant; Tukey's test; n = 3, average±s.d.
6.Extended Data Figure 6: Conversion of somatic tissue cells into STAP cells. (371 KB) a, Alkaline phosphatase expression of STAP cells derived from adipose-derived mesenchymal cells. Scale bar, 100μm. b, E-cadherin expression of STAP cells derived from adipose-derived mesenchymal cells. Scale bar, 50μm. c, FACS sorting of dissociated neonatal cardiac muscle cells by removing CD45+ cells. d, Cardiomyocyte marker gene expression during STAP conversion from cardiomyocytes (n = 3, average±s.d.).
7.Extended Data Figure 7: Generation chimaeras with STAP cells. (170 KB) a, 2N chimaeras generated with STAP cells derived from Oct4-gfp C57BL/6 mice (left) and 129/Sv×C57BL/6 F1 mice (right). b, Generation of chimaeric mice from STAP cells by cluster injection. STAP cells used in the experiments above were generated from CD45+ lymphocytes of multiple neonatal spleens (male and female tissues were mixed). *All fetuses were collected at 13.5d.p.c. to 15.5d.p.c. and the contribution rate of STAP cells into each organ was examined by FACS. **The contribution of STAP cells into each chimaera was scored as high (>50% of the coat colour of GFP expression). ***B6GFP: C57BL/6 mouse carrying cag-gfp.c, Production of offspring from STAP cells via germline transmission. Chimaeras generated with 129/Sv×B6GFP STAP cells (obtained from the experiments shown in b) were used for germline transmission study. d, 4N embryos at E9.5 generated with STAP cells derived from F1 GFP mice (B6GFP and DBA/2 or 129/Sv). B6GFP, C57BL/6 mouse carrying cag-gfp.
8.Extended Data Figure 8: Molecular and cellular characterization of STAP stem cells. (347 KB) a, Compatibility of 2i conditions with STAP stem-cell derivation from STAP cells and STAP stem-cell maintenance. STAP stem cells could not be established directly from STAP cells in 2i + LIF medium (top). However, once established in ACTH medium, STAP stem cells were able to survive and expand in 2i + LIF medium. Scale bar, 100μm. b, Q-band analysis (n = 4; all cell lines showed the normal karyotype). c, Multicolour FISH analysis (n = 8; all cell lines showed the normal karyotype) of STAP stem cells. d, Methylation status of the Oct4 and Nanog promoters. e, Electron microscope analysis of STAP stem cells. Scale bar, 1μm. f, g, Beating cardiac muscle (mesoderm; 38%, n = 8). Red line indicates an analysed region for kymograph (g). h, Clonability of STAP stem cells. Clonal expansion from single STAP stem cells was performed. Pluripotency of clonal cell lines was confirmed by teratoma formation assay, showing the formation of neuroectoderm (left), muscle tissue (middle) and bronchial-like epithelium (right). Scale bar, 100μm. i, Production of chimaeric mice from STAP stem-cell lines using diploid embryos. *These STAP stem-cell lines were generated from independent STAP cell clusters. j, Production of mouse chimaeras from STAP stem-cell lines by the tetraploid complementation method. *These STAP stem-cell lines were generated from independent STAP cell clusters. k, No H3K27me3-dense foci are seen in female STAP stem cells (n = 50; the CD45+ cell is a positive control). Scale bar, 10μm.
9.Extended Data Figure 9: Effects of various stressors on STAP conversion. (123 KB) a, Percentages of Oct4-GFP-expressing cells 7days after stress treatment. Somatic cells were isolated from various tissues and exposed to different stressors. Oct4-GFP expression was analysed by FACS. b, Oct4 and Oct4-GFP expression induced in the reflux oesophagitis mouse model as an in vivo acid exposure model (top, experimental procedure). Oct4, but not Nanog, expression was observed in the oesophageal epithelium exposed to gastric acid (75% of 12 operated mice).
Video Video 1: Live imaging of low-pH-treated CD45+cells (22.67 MB, Download)
DIC images during day 0 – day 7, overlaid with oct3/4::GFP (green). A strong contrast of DIC (as compared to video 2) was applied to imaging so that lamellipodia-like processes (frequently seen on and after day 4) could be viewed easily. Video 2: Live imaging of low-pH-treated CD45+cells (another view) (11.62 MB, Download) DIC images during day 0 – day 6, overlaid with oct3/4::GFP (green). The interval of imaging was half (15 min) of that of video 1 (the overall speed of the video is three-times slower than video 1). In this view field where the cell density was relatively low, behaviours of individual cells were more easily seen. In this case, forming clusters were slightly smaller in size.
Extended Data Figure 1: Conversion of haematopoietic cells into Oct4-GFP+ cells by a low-pH exposure.
Hover over figure to zoom
a, Optimization of pH conditions for Oct4-GFP induction. Five days after CD45-positive cells were exposed to acidic solution treatment at different pH, Oct4-GFP expression was analysed by FACS (n = 3, average±s.d.). b, Gating strategy for Oct4-GFP+ cell sorting. Top: representative results 7days after the stress treatment. Bottom: non-treated control. P3 populations were sorted and counted as Oct4-GFP+ cells for all experiments. c, Controls for FACS analysis. In Oct4-GFP+ cell analysis, the grey and white histograms indicate the negative control (non-stress-treated Oct4-gfp haematopoietic cells) and the positive control (Oct4-gfp ES cells), respectively. Also, the green histograms indicate non-treated cells (left) and stress-treated cells at day7 (right). In CD45+ cell analysis, the grey and white histograms indicate the negative (isotype) and positive controls, respectively. The red histograms indicate non-stress-treated cells (left) and stress-treated cells at day7 (right). d, Oct4-GFP+ cell generation from various subpopulations of CD45+ cells. Seven days after the stress treatment, Oct4-GFP expression was analysed by FACS (n = 3, average±s.d.). Among total CD45+ fraction and its subfractions of CD19+, CD90+, CD34+ and CD34− cells, the efficacy of CD34+ cells was significantly lower than the others. P<0.05 by the Newman–Keuls test and P<0.01 by one-way ANOVA. e, Comparison of culture conditions for low-pH-induced conversion. Stress-treated cells were cultured in various media. The number of Oct4-GFP-expressing clusters was counted at day 14 (n = 3, average±s.d.). ***P<0.001 (B27+LIF versus all other groups); Tukey’s test. In the case of 3i medium, although the clusters appeared at a moderate efficiency, they appeared late and grew slowly. ACTH, ACTH-containing ES medium; ES+LIF+FBS, 20% FBS+LIF-containing ES culture medium; B27, DMEM/F12 medium containing 2% B27; B27+LIF, DMEM/F12 medium containing 2% B27+LIF; EpiSC, EpiSC culture medium containing Fgf2+activin. f, Signalling factor dependency of STAP cell generation. Growth factors that are conventionally used for pluripotent cell culture such as LIF, activin, Bmp4 and Fgf2 were added to basal culture medium (B27-supplemented DMEM/F12) in different culture phases (days 0–7, 2–7 and 4–7), and Oct4-GFP expression was analysed by FACS at day 7 (n = 3, average±s.d.). g, h, Time course of apoptosis after the low-pH exposure. Stress-treated cells and non-stress-treated control cells were stained with CD45, annexin-V and propidium iodide at day 0 (immediately after stress treatment), day 3 and day 7. g, Blue bars, GFP+CD45−; orange bars, GFP−CD45+. Percentages in total cells included propidium-iodide-positive cells. h, Annexin-V-positive cells in these cell populations were analysed by FACS.
Extended Data Figure 2: Phenotypic change during STAP cell conversion.
Hover over figure to zoom
a, Oct4 protein expression in STAP cells was detected by immunostaining at day 2 (left) and day 7 (right). b, Live cell imaging of STAP conversion (grey, CD45 antibody; green, Oct4-GFP). See Methods for experimental details to monitor live CD45 immunostaining. c, Immunostaining of a parental CD45+ cell (left) and an Oct4-GFP+ cell (right). Scale bar, 10μm. d, EdU uptake assay (n = 3, average±s.d.). e, Schematic of Tcrb gene rearrangement. f, T-cell-derived STAP cells. Scale bar, 100μm. g, Genomic PCR analysis of (D)J recombination at the Tcrb gene of T-cell-derived STAP cells. G.L. is the size of the non-rearranged germline type, whereas the smaller ladders correspond to the alternative rearrangements of J exons (confirmed by sequencing). Negative controls (ES cells), positive controls (lymphocytes) and T-cell-derived STAP (two independent preparations on d7) are indicated.
Extended Data Figure 3: Gene expression analyses during STAP conversion and endoderm differentiation assay.
Hover over figure to zoom
a, Expression of pluripotency marker genes in STAP cells derived from T cells (n = 3, average±s.d.). b, Expression of pluripotency marker genes in STAP cells. In this experiment, Oct4-GFP+ cells seen in live cell imaging (Extended Data Fig. 2b) were analysed to confirm their conversion into STAP cells (n = 3, average±s.d.). c, Haematopoietic marker expression during STAP conversion from CD45+ cells (n = 3, average±s.d.). d, Formation of visceral endoderm-like surface epithelium in differentiating STAP cluster on day 2 (left) and day 8 (right). Scale bars, 50μm.
Extended Data Figure 4: Teratoma formation assay and characterization of Oct4-GFP-dim cells.
Hover over figure to zoom
a–c, Teratomas formed from STAP cell clusters included neuroepithelium (a), striated muscle (b) and pancreas (c; right, high-magnification view showing a typical acinar morphology and ductal structures). Scale bars, 100μm. d, Teratoma-forming ability of Oct4-GFP+ and Oct4-GFP-dim cells (isolated by FACS, top). Oct4-GFP+ cells, but not Oct4-GFP-dim cells, efficiently formed teratomas (table at the bottom). However, because STAP cells were dissociation-intolerant, the teratoma-forming efficiency of dissociated Oct4-GFP+ cells was lower than that of non-dissociated STAP cell clusters. e, Gene expression of Oct4-GFP+ and Oct4-GFP-dim cells (n = 3, the average±s.d.). Haematopoietic marker gene expression (left) and early lineage marker gene expression (right) are shown.
Extended Data Figure 5: In vitro characterization of STAP cells.
Hover over figure to zoom
a, Immunostaining for Ki67 and BrdU. STAP cell clusters (top) and ES cell colonies (bottom) are shown. For BrdU uptake, BrdU was added into each culture medium (10μM) for 12h until fixation. Scale bar, 100μm. b, Transformation assay by soft agar culture. Neither Oct4-GFP+ nor Oct4-GFP-dim cells showed colony formation in soft agar, whereas ES cells and STAP stem cells showed anchorage-independent growth in the same LIF-B27 medium. Scale bar, 100µm. Proliferated cells were lysed and the amount of DNA in each well was estimated by chemical luminescence (graph). n = 3 , average±s.d. c, No substantial change in chromosome number was seen with STAP cells in the CGH array. Genomic DNA derived from CD45+ cells (male) was used as reference DNA. The spikes (for example, those seen in the X chromosome) were nonspecific and also found in the data of these parental CD45+ cells when the manufacturer’s control DNA was used as a reference. d, qPCR analysis for pluripotency markers that highly express in ES cells, but not in EpiSCs. Average±s.d. e, Immunostaining of markers for mouse EpiSC and ES cells. Scale bar, 100 μm. f, g, H3K27me3+ foci in female cells, which are indicative of X-chromosomal inactivation. These foci were not observed in male cells. Scale bar, 10μm. In the case of female STAP cells, ~40% of cells retained H3K27me3+ foci (g). **P<0.001; Tukey’s test. n = 3, average±s.d. Although nuclear staining looked to be higher in STAP cells with H3K27me3+ foci (f), this appeared to be caused by some optical artefacts scattering from the strong focal signal. h, qPCR analysis for the tight junction markers Zo-1 and claudin 7, which were highly expressed in EpiSCs, but not in ES cells or STAP cells. **P<0.01; ns, not significant; Tukey's test; n = 3, average±s.d.
Extended Data Figure 7: Generation chimaeras with STAP cells.
Hover over figure to zoom
a, 2N chimaeras generated with STAP cells derived from Oct4-gfp C57BL/6 mice (left) and 129/Sv×C57BL/6 F1 mice (right). b, Generation of chimaeric mice from STAP cells by cluster injection. STAP cells used in the experiments above were generated from CD45+ lymphocytes of multiple neonatal spleens (male and female tissues were mixed). *All fetuses were collected at 13.5d.p.c. to 15.5d.p.c. and the contribution rate of STAP cells into each organ was examined by FACS. **The contribution of STAP cells into each chimaera was scored as high (>50% of the coat colour of GFP expression). ***B6GFP: C57BL/6 mouse carrying cag-gfp.c, Production of offspring from STAP cells via germline transmission. Chimaeras generated with 129/Sv×B6GFP STAP cells (obtained from the experiments shown in b) were used for germline transmission study. d, 4N embryos at E9.5 generated with STAP cells derived from F1 GFP mice (B6GFP and DBA/2 or 129/Sv). B6GFP, C57BL/6 mouse carrying cag-gfp.
Extended Data Figure 8: Molecular and cellular characterization of STAP stem cells.
Hover over figure to zoom
a, Compatibility of 2i conditions with STAP stem-cell derivation from STAP cells and STAP stem-cell maintenance. STAP stem cells could not be established directly from STAP cells in 2i + LIF medium (top). However, once established in ACTH medium, STAP stem cells were able to survive and expand in 2i + LIF medium. Scale bar, 100μm. b, Q-band analysis (n = 4; all cell lines showed the normal karyotype). c, Multicolour FISH analysis (n = 8; all cell lines showed the normal karyotype) of STAP stem cells. d, Methylation status of the Oct4 and Nanog promoters. e, Electron microscope analysis of STAP stem cells. Scale bar, 1μm. f, g, Beating cardiac muscle (mesoderm; 38%, n = 8). Red line indicates an analysed region for kymograph (g). h, Clonability of STAP stem cells. Clonal expansion from single STAP stem cells was performed. Pluripotency of clonal cell lines was confirmed by teratoma formation assay, showing the formation of neuroectoderm (left), muscle tissue (middle) and bronchial-like epithelium (right). Scale bar, 100μm. i, Production of chimaeric mice from STAP stem-cell lines using diploid embryos. *These STAP stem-cell lines were generated from independent STAP cell clusters. j, Production of mouse chimaeras from STAP stem-cell lines by the tetraploid complementation method. *These STAP stem-cell lines were generated from independent STAP cell clusters. k, No H3K27me3-dense foci are seen in female STAP stem cells (n = 50; the CD45+ cell is a positive control). Scale bar, 10μm.
Extended Data Figure 9: Effects of various stressors on STAP conversion.
Hover over figure to zoom
a, Percentages of Oct4-GFP-expressing cells 7days after stress treatment. Somatic cells were isolated from various tissues and exposed to different stressors. Oct4-GFP expression was analysed by FACS. b, Oct4 and Oct4-GFP expression induced in the reflux oesophagitis mouse model as an in vivo acid exposure model (top, experimental procedure). Oct4, but not Nanog, expression was observed in the oesophageal epithelium exposed to gastric acid (75% of 12 operated mice).
Video 1: Live imaging of low-pH-treated CD45+cells
DIC images during day 0 – day 7, overlaid with oct3/4::GFP (green). A strong contrast of DIC (as compared to video 2) was applied to imaging so that lamellipodia-like processes (frequently seen on and after day 4) could be viewed easily.
Video 2: Live imaging of low-pH-treated CD45+cells (another view)
DIC images during day 0 – day 6, overlaid with oct3/4::GFP (green). The interval of imaging was half (15 min) of that of video 1 (the overall speed of the video is three-times slower than video 1). In this view field where the cell density was relatively low, behaviours of individual cells were more easily seen. In this case, forming clusters were slightly smaller in size.