2018年10月29日星期一

The Nobel Prize in Physiology or Medicine 1999

The Nobel Prize in Physiology or Medicine 1999 was awarded to Günter Blobel "for the discovery that proteins have intrinsic signals that govern their transport and localization in the cell."

Günter Blobel Facts:

Günter Blobel

Born: 21 May 1936, Waltersdorf (now Niegoslawice), Germany (now Poland)

Died: 18 February 2018, New York, NY, USA

Affiliation at the time of the award: Rockefeller University, New York, NY, USA

Summary
A large number of proteins carrying out essential functions are constantly being made within our cells. These proteins have to be transported either out of the cell, or to the different compartments – the organelles – within the cell. How are newly made proteins transported across the membrane surrounding the organelles, and how are they directed to their correct location?

These questions have been answered through the work of this year’s Nobel Laureate in Physiology or Medicine, Dr Günter Blobel, a cell and molecular biologist at the Rockefeller University in New York. Already at the beginning of the 1970s he discovered that newly synthesized proteins have an intrinsic signal that is essential for governing them to and across the membrane of the endoplasmic reticulum, one of the cell’s organelles. During the next twenty years Blobel characterized in detail the molecular mechanisms underlying these processes. He also showed that similar “address tags”, or “zip codes”, direct proteins to other intracellular organelles.

The principles discovered and described by Günter Blobel turned out to be universal, operating similarly in yeast, plant, and animal cells. A number of human hereditary diseases are caused by errors in these signals and transport mechanisms. Blobel’s research has also contributed to the development of a more effective use of cells as “protein factories” for the production of important drugs.



Please click https://www.nobelprize.org/prizes/medicine/1999/press-release/ for more detailed information.

 

The Nobel Prize in Physiology or Medicine 1999

The Nobel Prize in Physiology or Medicine 1999 was awarded to Günter Blobel "for the discovery that proteins have intrinsic signals that govern their transport and localization in the cell."

Günter Blobel Facts:

Günter Blobel

Born: 21 May 1936, Waltersdorf (now Niegoslawice), Germany (now Poland)

Died: 18 February 2018, New York, NY, USA

Affiliation at the time of the award: Rockefeller University, New York, NY, USA

Summary
A large number of proteins carrying out essential functions are constantly being made within our cells. These proteins have to be transported either out of the cell, or to the different compartments – the organelles – within the cell. How are newly made proteins transported across the membrane surrounding the organelles, and how are they directed to their correct location?

These questions have been answered through the work of this year’s Nobel Laureate in Physiology or Medicine, Dr Günter Blobel, a cell and molecular biologist at the Rockefeller University in New York. Already at the beginning of the 1970s he discovered that newly synthesized proteins have an intrinsic signal that is essential for governing them to and across the membrane of the endoplasmic reticulum, one of the cell’s organelles. During the next twenty years Blobel characterized in detail the molecular mechanisms underlying these processes. He also showed that similar “address tags”, or “zip codes”, direct proteins to other intracellular organelles.

The principles discovered and described by Günter Blobel turned out to be universal, operating similarly in yeast, plant, and animal cells. A number of human hereditary diseases are caused by errors in these signals and transport mechanisms. Blobel’s research has also contributed to the development of a more effective use of cells as “protein factories” for the production of important drugs.



Please click https://www.nobelprize.org/prizes/medicine/1999/press-release/ for more detailed information.

 

2018年10月26日星期五

The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation

Content introduction:

  • A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans

  • In utero priming of highly functional effector T cell responses to human malaria

  • Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

  • Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling

  • The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation


1. A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans

Periodontitis is one of the most common human inflammatory diseases, yet the mechanisms that drive immunopathology and could be therapeutically targeted are not well defined. Here, Nicolas Dutzan at NIH in Bethesda, USA and his colleagues demonstrate an expansion of resident memory T helper 17 (TH17) cells in human periodontitis. Phenocopying humans, TH17 cells expanded in murine experimental periodontitis through local proliferation. Unlike homeostatic oral TH17 cells, which accumulate in a commensal-independent and interleukin-6 (IL-6)–dependent manner, periodontitis-associated expansion of TH17 cells was dependent on the local dysbiotic microbiome and required both IL-6 and IL-23. TH17 cells and associated neutrophil accumulation were necessary for inflammatory tissue destruction in experimental periodontitis. Genetic or pharmacological inhibition of TH17 cell differentiation conferred protection from immunopathology. Studies in a unique patient population with a genetic defect in TH17 cell differentiation established human relevance for our murine experimental studies. In the oral cavity, human TH17 cell defects were associated with diminished periodontal inflammation and bone loss, despite increased prevalence of recurrent oral fungal infections. Their study highlights distinct functions of TH17 cells in oral immunity and inflammation and paves the way to a new targeted therapeutic approach for the treatment of periodontitis.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat0797

2. In utero priming of highly functional effector T cell responses to human malaria

Malaria remains a significant cause of morbidity and mortality worldwide, particularly in infants and children. Some studies have reported that exposure to malaria antigens in utero results in the development of tolerance, which could contribute to poor immunity to malaria in early life. However, the effector T cell response to pathogen-derived antigens encountered in utero, including malaria, has not been well characterized. Here, Pamela M. Odorizzi at University of California in San Francisco, USA and his colleagues assessed the frequency, phenotype, and function of cord blood T cells from Ugandan infants born to mothers with and without placental malaria. They found that infants born to mothers with active placental malaria had elevated frequencies of proliferating effector memory fetal CD4+ T cells and higher frequencies of CD4+ and CD8+ T cells that produced inflammatory cytokines. Fetal CD4+ and CD8+ T cells from placental malaria–exposed infants exhibited greater in vitro proliferation to malaria antigens. Malaria-specific CD4+ T cell proliferation correlated with prospective protection from malaria during childhood. These data demonstrate that placental malaria is associated with the generation of proinflammatory malaria-responsive fetal T cells. These findings add to our current understanding of fetal immunity and indicate that a functional and protective pathogen-specific T cell response can be generated in utero.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat6176

3. Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

A primary barrier to the success of T cell–recruiting bispecific antibodies in the treatment of solid tumors is the lack of tumor-specific targets, resulting in on-target off-tumor adverse effects from T cell autoreactivity to target-expressing organs. To overcome this, Dionysos Slaga at Genentech Inc in South San Francisco, USA and his colleagues developed an anti-HER2/CD3 T cell–dependent bispecific (TDB) antibody that selectively targets HER2-overexpressing tumor cells with high potency, while sparing cells that express low amounts of HER2 found in normal human tissues. Selectivity is based on the avidity of two low-affinity anti-HER2 Fab arms to high target density on HER2-overexpressing cells. The increased selectivity to HER2-overexpressing cells is expected to mitigate the risk of adverse effects and increase the therapeutic index. Results included in this manuscript not only support the clinical development of anti-HER2/CD3 1Fab–immunoglobulin G TDB but also introduce a potentially widely applicable strategy for other T cell–directed therapies. The potential of this discovery has broad applications to further enable consideration of solid tumor targets that were previously limited by on-target, but off-tumor, autoimmunity.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat5775

4. Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling

In many mammals, the eyelids migrate over the eye and fuse during embryogenesis to protect the cornea from damage during birth and early life. Loss-of-function mutations affecting the epidermal growth factor receptor (EGFR) signaling pathway cause an eyes-open-at-birth (EOB) phenotype in rodents. Ganlan Bian at Fourth Military Medical University in Xi’an, China and his colleagues identified an insertional mutation in Spinster homolog 2 (Spns2) in a strain of transgenic rats exhibiting the EOB phenotype. Spns2, a sphingosine 1-phosphate (S1P) transporter that releases S1P from cells, was enriched at the tip of developing eyelids in wild-type rat embryos. Spns2 expression or treatment with S1P or any one of several EGFR ligands rescued the EOB Spns2 mutant phenotype in vivo and in tissue explants in vitro and rescued the formation of stress fibers in primary keratinocytes from mutants. S1P signaled through the receptors S1PR1, S1PR2, and S1PR3 to activate extracellular signal–regulated kinase (ERK) and EGFR-dependent mitogen-activated protein kinase kinase kinase 1 (MEKK1)–c-Jun signaling. S1P also induced the nuclear translocation of the transcription factor MAL in a manner dependent on EGFR signaling. MAL and c-Jun stimulated the expression of the microRNAs miR-21 and miR-222, both of which target the metalloprotease inhibitor TIMP3, thus promoting metalloprotease activity. The metalloproteases ADAM10 and ADAM17 stimulated EGFR signaling by cleaving a membrane-anchored form of EGF to release the ligand. Their results outline a network by which S1P transactivates EGFR signaling through a complex mechanism involving feedback between several intra- and extracellular molecules to promote eyelid fusion in the developing rat.

Read more, please click http://stke.sciencemag.org/content/11/553/eaat1470

5. The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation

Fgr is a member of the Src family of nonreceptor tyrosine kinases, which are overexpressed and constitutively active in many human cancers. Fgr expression is restricted to myeloid hematopoietic cells and is markedly increased in a subset of bone marrow samples from patients with acute myeloid leukemia (AML). Here, Kexin Shen at University of Pittsburgh School of Medicine in Pittsburgh, USA and his colleagues investigated the oncogenic potential of Fgr using Rat-2 fibroblasts that do not express the kinase. Expression of either wild-type or regulatory tail-mutant constructs of Fgr promoted cellular transformation (inferred from colony formation in soft agar), which was accompanied by phosphorylation of the Fgr activation loop, suggesting that the kinase domain of Fgr functions independently of regulation by its noncatalytic SH3-SH2 region. Unlike other family members, recombinant Fgr was not activated by SH3-SH2 domain ligands. However, hydrogen-deuterium exchange mass spectrometry data suggested that the regulatory SH3 and SH2 domains packed against the back of the kinase domain in a Src-like manner. Sequence alignment showed that the activation loop of Fgr was distinct from that of all other Src family members, with proline rather than alanine at the +2 position relative to the activation loop tyrosine. Substitution of the activation loop of Fgr with the sequence from Src partially inhibited kinase activity and suppressed colony formation. Last, Fgr expression enhanced the sensitivity of human myeloid progenitor cells to the cytokine GM-CSF. Because its kinase domain is not sensitive to SH3-SH2–mediated control, simple overexpression of Fgr without mutation may contribute to oncogenic transformation in AML and other blood cancers.



Read more, please click http://stke.sciencemag.org/content/11/553/eaat5916

2018年10月25日星期四

The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation

Content introduction:

  • A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans

  • In utero priming of highly functional effector T cell responses to human malaria

  • Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

  • Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling

  • The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation


1. A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans

Periodontitis is one of the most common human inflammatory diseases, yet the mechanisms that drive immunopathology and could be therapeutically targeted are not well defined. Here, Nicolas Dutzan at NIH in Bethesda, USA and his colleagues demonstrate an expansion of resident memory T helper 17 (TH17) cells in human periodontitis. Phenocopying humans, TH17 cells expanded in murine experimental periodontitis through local proliferation. Unlike homeostatic oral TH17 cells, which accumulate in a commensal-independent and interleukin-6 (IL-6)–dependent manner, periodontitis-associated expansion of TH17 cells was dependent on the local dysbiotic microbiome and required both IL-6 and IL-23. TH17 cells and associated neutrophil accumulation were necessary for inflammatory tissue destruction in experimental periodontitis. Genetic or pharmacological inhibition of TH17 cell differentiation conferred protection from immunopathology. Studies in a unique patient population with a genetic defect in TH17 cell differentiation established human relevance for our murine experimental studies. In the oral cavity, human TH17 cell defects were associated with diminished periodontal inflammation and bone loss, despite increased prevalence of recurrent oral fungal infections. Their study highlights distinct functions of TH17 cells in oral immunity and inflammation and paves the way to a new targeted therapeutic approach for the treatment of periodontitis.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat0797

2. In utero priming of highly functional effector T cell responses to human malaria

Malaria remains a significant cause of morbidity and mortality worldwide, particularly in infants and children. Some studies have reported that exposure to malaria antigens in utero results in the development of tolerance, which could contribute to poor immunity to malaria in early life. However, the effector T cell response to pathogen-derived antigens encountered in utero, including malaria, has not been well characterized. Here, Pamela M. Odorizzi at University of California in San Francisco, USA and his colleagues assessed the frequency, phenotype, and function of cord blood T cells from Ugandan infants born to mothers with and without placental malaria. They found that infants born to mothers with active placental malaria had elevated frequencies of proliferating effector memory fetal CD4+ T cells and higher frequencies of CD4+ and CD8+ T cells that produced inflammatory cytokines. Fetal CD4+ and CD8+ T cells from placental malaria–exposed infants exhibited greater in vitro proliferation to malaria antigens. Malaria-specific CD4+ T cell proliferation correlated with prospective protection from malaria during childhood. These data demonstrate that placental malaria is associated with the generation of proinflammatory malaria-responsive fetal T cells. These findings add to our current understanding of fetal immunity and indicate that a functional and protective pathogen-specific T cell response can be generated in utero.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat6176

3. Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

A primary barrier to the success of T cell–recruiting bispecific antibodies in the treatment of solid tumors is the lack of tumor-specific targets, resulting in on-target off-tumor adverse effects from T cell autoreactivity to target-expressing organs. To overcome this, Dionysos Slaga at Genentech Inc in South San Francisco, USA and his colleagues developed an anti-HER2/CD3 T cell–dependent bispecific (TDB) antibody that selectively targets HER2-overexpressing tumor cells with high potency, while sparing cells that express low amounts of HER2 found in normal human tissues. Selectivity is based on the avidity of two low-affinity anti-HER2 Fab arms to high target density on HER2-overexpressing cells. The increased selectivity to HER2-overexpressing cells is expected to mitigate the risk of adverse effects and increase the therapeutic index. Results included in this manuscript not only support the clinical development of anti-HER2/CD3 1Fab–immunoglobulin G TDB but also introduce a potentially widely applicable strategy for other T cell–directed therapies. The potential of this discovery has broad applications to further enable consideration of solid tumor targets that were previously limited by on-target, but off-tumor, autoimmunity.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat5775

4. Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling

In many mammals, the eyelids migrate over the eye and fuse during embryogenesis to protect the cornea from damage during birth and early life. Loss-of-function mutations affecting the epidermal growth factor receptor (EGFR) signaling pathway cause an eyes-open-at-birth (EOB) phenotype in rodents. Ganlan Bian at Fourth Military Medical University in Xi’an, China and his colleagues identified an insertional mutation in Spinster homolog 2 (Spns2) in a strain of transgenic rats exhibiting the EOB phenotype. Spns2, a sphingosine 1-phosphate (S1P) transporter that releases S1P from cells, was enriched at the tip of developing eyelids in wild-type rat embryos. Spns2 expression or treatment with S1P or any one of several EGFR ligands rescued the EOB Spns2 mutant phenotype in vivo and in tissue explants in vitro and rescued the formation of stress fibers in primary keratinocytes from mutants. S1P signaled through the receptors S1PR1, S1PR2, and S1PR3 to activate extracellular signal–regulated kinase (ERK) and EGFR-dependent mitogen-activated protein kinase kinase kinase 1 (MEKK1)–c-Jun signaling. S1P also induced the nuclear translocation of the transcription factor MAL in a manner dependent on EGFR signaling. MAL and c-Jun stimulated the expression of the microRNAs miR-21 and miR-222, both of which target the metalloprotease inhibitor TIMP3, thus promoting metalloprotease activity. The metalloproteases ADAM10 and ADAM17 stimulated EGFR signaling by cleaving a membrane-anchored form of EGF to release the ligand. Their results outline a network by which S1P transactivates EGFR signaling through a complex mechanism involving feedback between several intra- and extracellular molecules to promote eyelid fusion in the developing rat.

Read more, please click http://stke.sciencemag.org/content/11/553/eaat1470

5. The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation

Fgr is a member of the Src family of nonreceptor tyrosine kinases, which are overexpressed and constitutively active in many human cancers. Fgr expression is restricted to myeloid hematopoietic cells and is markedly increased in a subset of bone marrow samples from patients with acute myeloid leukemia (AML). Here, Kexin Shen at University of Pittsburgh School of Medicine in Pittsburgh, USA and his colleagues investigated the oncogenic potential of Fgr using Rat-2 fibroblasts that do not express the kinase. Expression of either wild-type or regulatory tail-mutant constructs of Fgr promoted cellular transformation (inferred from colony formation in soft agar), which was accompanied by phosphorylation of the Fgr activation loop, suggesting that the kinase domain of Fgr functions independently of regulation by its noncatalytic SH3-SH2 region. Unlike other family members, recombinant Fgr was not activated by SH3-SH2 domain ligands. However, hydrogen-deuterium exchange mass spectrometry data suggested that the regulatory SH3 and SH2 domains packed against the back of the kinase domain in a Src-like manner. Sequence alignment showed that the activation loop of Fgr was distinct from that of all other Src family members, with proline rather than alanine at the +2 position relative to the activation loop tyrosine. Substitution of the activation loop of Fgr with the sequence from Src partially inhibited kinase activity and suppressed colony formation. Last, Fgr expression enhanced the sensitivity of human myeloid progenitor cells to the cytokine GM-CSF. Because its kinase domain is not sensitive to SH3-SH2–mediated control, simple overexpression of Fgr without mutation may contribute to oncogenic transformation in AML and other blood cancers.



Read more, please click http://stke.sciencemag.org/content/11/553/eaat5916

The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation

Content introduction:

  • A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans

  • In utero priming of highly functional effector T cell responses to human malaria

  • Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

  • Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling

  • The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation


1. A dysbiotic microbiome triggers TH17 cells to mediate oral mucosal immunopathology in mice and humans

Periodontitis is one of the most common human inflammatory diseases, yet the mechanisms that drive immunopathology and could be therapeutically targeted are not well defined. Here, Nicolas Dutzan at NIH in Bethesda, USA and his colleagues demonstrate an expansion of resident memory T helper 17 (TH17) cells in human periodontitis. Phenocopying humans, TH17 cells expanded in murine experimental periodontitis through local proliferation. Unlike homeostatic oral TH17 cells, which accumulate in a commensal-independent and interleukin-6 (IL-6)–dependent manner, periodontitis-associated expansion of TH17 cells was dependent on the local dysbiotic microbiome and required both IL-6 and IL-23. TH17 cells and associated neutrophil accumulation were necessary for inflammatory tissue destruction in experimental periodontitis. Genetic or pharmacological inhibition of TH17 cell differentiation conferred protection from immunopathology. Studies in a unique patient population with a genetic defect in TH17 cell differentiation established human relevance for our murine experimental studies. In the oral cavity, human TH17 cell defects were associated with diminished periodontal inflammation and bone loss, despite increased prevalence of recurrent oral fungal infections. Their study highlights distinct functions of TH17 cells in oral immunity and inflammation and paves the way to a new targeted therapeutic approach for the treatment of periodontitis.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat0797

2. In utero priming of highly functional effector T cell responses to human malaria

Malaria remains a significant cause of morbidity and mortality worldwide, particularly in infants and children. Some studies have reported that exposure to malaria antigens in utero results in the development of tolerance, which could contribute to poor immunity to malaria in early life. However, the effector T cell response to pathogen-derived antigens encountered in utero, including malaria, has not been well characterized. Here, Pamela M. Odorizzi at University of California in San Francisco, USA and his colleagues assessed the frequency, phenotype, and function of cord blood T cells from Ugandan infants born to mothers with and without placental malaria. They found that infants born to mothers with active placental malaria had elevated frequencies of proliferating effector memory fetal CD4+ T cells and higher frequencies of CD4+ and CD8+ T cells that produced inflammatory cytokines. Fetal CD4+ and CD8+ T cells from placental malaria–exposed infants exhibited greater in vitro proliferation to malaria antigens. Malaria-specific CD4+ T cell proliferation correlated with prospective protection from malaria during childhood. These data demonstrate that placental malaria is associated with the generation of proinflammatory malaria-responsive fetal T cells. These findings add to our current understanding of fetal immunity and indicate that a functional and protective pathogen-specific T cell response can be generated in utero.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat6176

3. Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3

A primary barrier to the success of T cell–recruiting bispecific antibodies in the treatment of solid tumors is the lack of tumor-specific targets, resulting in on-target off-tumor adverse effects from T cell autoreactivity to target-expressing organs. To overcome this, Dionysos Slaga at Genentech Inc in South San Francisco, USA and his colleagues developed an anti-HER2/CD3 T cell–dependent bispecific (TDB) antibody that selectively targets HER2-overexpressing tumor cells with high potency, while sparing cells that express low amounts of HER2 found in normal human tissues. Selectivity is based on the avidity of two low-affinity anti-HER2 Fab arms to high target density on HER2-overexpressing cells. The increased selectivity to HER2-overexpressing cells is expected to mitigate the risk of adverse effects and increase the therapeutic index. Results included in this manuscript not only support the clinical development of anti-HER2/CD3 1Fab–immunoglobulin G TDB but also introduce a potentially widely applicable strategy for other T cell–directed therapies. The potential of this discovery has broad applications to further enable consideration of solid tumor targets that were previously limited by on-target, but off-tumor, autoimmunity.

Read more, please click http://stm.sciencemag.org/content/10/463/eaat5775

4. Sphingosine 1-phosphate stimulates eyelid closure in the developing rat by stimulating EGFR signaling

In many mammals, the eyelids migrate over the eye and fuse during embryogenesis to protect the cornea from damage during birth and early life. Loss-of-function mutations affecting the epidermal growth factor receptor (EGFR) signaling pathway cause an eyes-open-at-birth (EOB) phenotype in rodents. Ganlan Bian at Fourth Military Medical University in Xi’an, China and his colleagues identified an insertional mutation in Spinster homolog 2 (Spns2) in a strain of transgenic rats exhibiting the EOB phenotype. Spns2, a sphingosine 1-phosphate (S1P) transporter that releases S1P from cells, was enriched at the tip of developing eyelids in wild-type rat embryos. Spns2 expression or treatment with S1P or any one of several EGFR ligands rescued the EOB Spns2 mutant phenotype in vivo and in tissue explants in vitro and rescued the formation of stress fibers in primary keratinocytes from mutants. S1P signaled through the receptors S1PR1, S1PR2, and S1PR3 to activate extracellular signal–regulated kinase (ERK) and EGFR-dependent mitogen-activated protein kinase kinase kinase 1 (MEKK1)–c-Jun signaling. S1P also induced the nuclear translocation of the transcription factor MAL in a manner dependent on EGFR signaling. MAL and c-Jun stimulated the expression of the microRNAs miR-21 and miR-222, both of which target the metalloprotease inhibitor TIMP3, thus promoting metalloprotease activity. The metalloproteases ADAM10 and ADAM17 stimulated EGFR signaling by cleaving a membrane-anchored form of EGF to release the ligand. Their results outline a network by which S1P transactivates EGFR signaling through a complex mechanism involving feedback between several intra- and extracellular molecules to promote eyelid fusion in the developing rat.

Read more, please click http://stke.sciencemag.org/content/11/553/eaat1470

5. The Src family kinase Fgr is a transforming oncoprotein that functions independently of SH3-SH2 domain regulation

Fgr is a member of the Src family of nonreceptor tyrosine kinases, which are overexpressed and constitutively active in many human cancers. Fgr expression is restricted to myeloid hematopoietic cells and is markedly increased in a subset of bone marrow samples from patients with acute myeloid leukemia (AML). Here, Kexin Shen at University of Pittsburgh School of Medicine in Pittsburgh, USA and his colleagues investigated the oncogenic potential of Fgr using Rat-2 fibroblasts that do not express the kinase. Expression of either wild-type or regulatory tail-mutant constructs of Fgr promoted cellular transformation (inferred from colony formation in soft agar), which was accompanied by phosphorylation of the Fgr activation loop, suggesting that the kinase domain of Fgr functions independently of regulation by its noncatalytic SH3-SH2 region. Unlike other family members, recombinant Fgr was not activated by SH3-SH2 domain ligands. However, hydrogen-deuterium exchange mass spectrometry data suggested that the regulatory SH3 and SH2 domains packed against the back of the kinase domain in a Src-like manner. Sequence alignment showed that the activation loop of Fgr was distinct from that of all other Src family members, with proline rather than alanine at the +2 position relative to the activation loop tyrosine. Substitution of the activation loop of Fgr with the sequence from Src partially inhibited kinase activity and suppressed colony formation. Last, Fgr expression enhanced the sensitivity of human myeloid progenitor cells to the cytokine GM-CSF. Because its kinase domain is not sensitive to SH3-SH2–mediated control, simple overexpression of Fgr without mutation may contribute to oncogenic transformation in AML and other blood cancers.



Read more, please click http://stke.sciencemag.org/content/11/553/eaat5916

2018年10月22日星期一

The Nobel Prize in Physiology or Medicine 2000

The Nobel Prize in Physiology or Medicine 2000 was awarded jointly to Arvid Carlsson, Paul Greengard and Eric R. Kandel "for their discoveries concerning signal transduction in the nervous system."

















NobelistBornDiedAffiliation at the time of the award
Arvid Carlsson25 January 1923, Uppsala, Sweden29 June 2018, Gothenburg, SwedenGöteborg University, Gothenburg, Sweden
Paul Greengard11 December 1925, New York, NY, USARockefeller University, New York, NY, USA
Eric R. Kandel7 November 1929, Vienna, AustriaColumbia University, New York, NY, USA
Summary

In the human brain there are more than hundred billion nerve cells. They are connected to each other through an infinitely complex network of nerve processes. The message from one nerve cell to another is transmitted through different chemical transmitters. The signal transduction takes place in special points of contact, called synapses. A nerve cell can have thousands of such contacts with other nerve cells.

The three Nobel Laureates in Physiology or Medicine have made pioneering discoveries concerning one type of signal transduction between nerve cells, referred to as slow synaptic transmission. These discoveries have been crucial for an understanding of the normal function of the brain and how disturbances in this signal transduction can give rise to neurological and psychiatric diseases. These findings have resulted in the development of new drugs.



More details, please click The Nobel Prize in Physiology or Medicine 2000.

2018年10月21日星期日

The Nobel Prize in Physiology or Medicine 2000

The Nobel Prize in Physiology or Medicine 2000 was awarded jointly to Arvid Carlsson, Paul Greengard and Eric R. Kandel "for their discoveries concerning signal transduction in the nervous system."

















NobelistBornDiedAffiliation at the time of the award
Arvid Carlsson25 January 1923, Uppsala, Sweden29 June 2018, Gothenburg, SwedenGöteborg University, Gothenburg, Sweden
Paul Greengard11 December 1925, New York, NY, USARockefeller University, New York, NY, USA
Eric R. Kandel7 November 1929, Vienna, AustriaColumbia University, New York, NY, USA
Summary

In the human brain there are more than hundred billion nerve cells. They are connected to each other through an infinitely complex network of nerve processes. The message from one nerve cell to another is transmitted through different chemical transmitters. The signal transduction takes place in special points of contact, called synapses. A nerve cell can have thousands of such contacts with other nerve cells.

The three Nobel Laureates in Physiology or Medicine have made pioneering discoveries concerning one type of signal transduction between nerve cells, referred to as slow synaptic transmission. These discoveries have been crucial for an understanding of the normal function of the brain and how disturbances in this signal transduction can give rise to neurological and psychiatric diseases. These findings have resulted in the development of new drugs.



More details, please click The Nobel Prize in Physiology or Medicine 2000.

2018年10月19日星期五

Complement C5a Fosters Squamous Carcinogenesis and Limits T Cell Response to Chemotherapy

Content introduction:

  • Complement C5a Fosters Squamous Carcinogenesis and Limits T Cell Response to Chemotherapy

  • Enhanced Anti-lymphoma Activity of CAR19-iNKT Cells Underpinned by Dual CD19 and CD1d Targeting

  • Aberrant ERBB4-SRC Signaling as a Hallmark of Group 4 Medulloblastoma Revealed by Integrative Phosphoproteomic Profiling

  • Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups

  • The Oncogenic Transcription Factor RUNX1/ETO Corrupts Cell Cycle Regulation to Drive Leukemic Transformation


1. Complement C5a Fosters Squamous Carcinogenesis and Limits T Cell Response to Chemotherapy

Complement is a critical component of humoral immunity implicated in cancer development; however, its biological contributions to tumorigenesis remain poorly understood. Using the K14-HPV16 transgenic mouse model of squamous carcinogenesis, Terry R. Medler at Oregon Health & Science University in Portland, USA and his colleagues report that urokinase (uPA)+ macrophages regulate C3-independent release of C5a during premalignant progression, which in turn regulates protumorigenic properties of C5aR1+ mast cells and macrophages, including suppression of CD8+ T cell cytotoxicity. Therapeutic inhibition of C5aR1 via the peptide antagonist PMX-53 improved efficacy of paclitaxel chemotherapy associated with increased presence and cytotoxic properties of CXCR3+ effector memory CD8+ T cells in carcinomas, dependent on both macrophage transcriptional programming and IFNγ. Together, these data identify C5aR1-dependent signaling as an important immunomodulatory program in neoplastic tissue tractable for combinatorial cancer immunotherapy.



Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30417-3

2. Enhanced Anti-lymphoma Activity of CAR19-iNKT Cells Underpinned by Dual CD19 and CD1d Targeting

Chimeric antigen receptor anti-CD19 (CAR19)-T cell immunotherapy-induced clinical remissions in CD19+ B cell lymphomas are often short lived. Antonia Rotolo at Imperial College in London, UK and his colleagues tested whether CAR19-engineering of the CD1d-restricted invariant natural killer T (iNKT) cells would result in enhanced anti-lymphoma activity. CAR19-iNKT cells co-operatively activated by CD1d- and CAR19-CD19-dependent interactions are more effective than CAR19-T cells against CD1d-expressing lymphomas in vitro and in vivo. The swifter in vivo anti-lymphoma activity of CAR19-iNKT cells and their enhanced ability to eradicate brain lymphomas underpinned an improved tumor-free and overall survival. CD1D transcriptional de-repression by all-trans retinoic acid results in further enhanced cytotoxicity of CAR19-iNKT cells against CD19+ chronic lymphocytic leukemia cells. Thus, iNKT cells are a highly efficient platform for CAR-based immunotherapy of lymphomas and possibly other CD1d-expressing cancers.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30377-5

3. Aberrant ERBB4-SRC Signaling as a Hallmark of Group 4 Medulloblastoma Revealed by Integrative Phosphoproteomic Profiling

The current consensus recognizes four main medulloblastoma subgroups (wingless, Sonic hedgehog, group 3 and group 4). While medulloblastoma subgroups have been characterized extensively at the (epi-)genomic and transcriptomic levels, the proteome and phosphoproteome landscape remain to be comprehensively elucidated. Using quantitative (phospho)-proteomics in primary human medulloblastomas, Antoine Forget at Institut Curie, PSL Research University in Orsay, France and his colleagues unravel distinct posttranscriptional regulation leading to highly divergent oncogenic signaling and kinase activity profiles in groups 3 and 4 medulloblastomas. Specifically, proteomic and phosphoproteomic analyses identify aberrant ERBB4-SRC signaling in group 4. Hence, enforced expression of an activated SRC combined with p53 inactivation induces murine tumors that resemble group 4 medulloblastoma. Therefore, Their integrative proteogenomics approach unveils an oncogenic pathway and potential therapeutic vulnerability in the most common medulloblastoma subgroup.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30356-8

4. Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups

There is a pressing need to identify therapeutic targets in tumors with low mutation rates such as the malignant pediatric brain tumor medulloblastoma. To address this challenge, Tenley C. Archer at Boston Children's Hospital in Boston, USA and his colleagues quantitatively profiled global proteomes and phospho-proteomes of 45 medulloblastoma samples. Integrated analyses revealed that tumors with similar RNA expression vary extensively at the post-transcriptional and post-translational levels. They identified distinct pathways associated with two subsets of SHH tumors, and found post-translational modifications of MYC that are associated with poor outcomes in group 3 tumors. They found kinases associated with subtypes and showed that inhibiting PRKDC sensitizes MYC-driven cells to radiation. Their study shows that proteomics enables a more comprehensive, functional readout, providing a foundation for future therapeutic strategies.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30358-1

5. The Oncogenic Transcription Factor RUNX1/ETO Corrupts Cell Cycle Regulation to Drive Leukemic Transformation

Oncogenic transcription factors such as the leukemic fusion protein RUNX1/ETO, which drives t(8;21) acute myeloid leukemia (AML), constitute cancer-specific but highly challenging therapeutic targets. Natalia Martinez-Soria at Newcastle University in Newcastle upon Tyne, UK and his colleagues used epigenomic profiling data for an RNAi screen to interrogate the transcriptional network maintaining t(8;21) AML. This strategy identified Cyclin D2 (CCND2) as a crucial transmitter of RUNX1/ETO-driven leukemic propagation. RUNX1/ETO cooperates with AP-1 to drive CCND2 expression. Knockdown or pharmacological inhibition of CCND2 by an approved drug significantly impairs leukemic expansion of patient-derived AML cells and engraftment in immunodeficient murine hosts. Their data demonstrate that RUNX1/ETO maintains leukemia by promoting cell cycle progression and identifies G1 CCND-CDK complexes as promising therapeutic targets for treatment of RUNX1/ETO-driven AML.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30375-1

2018年10月18日星期四

Complement C5a Fosters Squamous Carcinogenesis and Limits T Cell Response to Chemotherapy

Content introduction:

  • Complement C5a Fosters Squamous Carcinogenesis and Limits T Cell Response to Chemotherapy

  • Enhanced Anti-lymphoma Activity of CAR19-iNKT Cells Underpinned by Dual CD19 and CD1d Targeting

  • Aberrant ERBB4-SRC Signaling as a Hallmark of Group 4 Medulloblastoma Revealed by Integrative Phosphoproteomic Profiling

  • Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups

  • The Oncogenic Transcription Factor RUNX1/ETO Corrupts Cell Cycle Regulation to Drive Leukemic Transformation


1. Complement C5a Fosters Squamous Carcinogenesis and Limits T Cell Response to Chemotherapy

Complement is a critical component of humoral immunity implicated in cancer development; however, its biological contributions to tumorigenesis remain poorly understood. Using the K14-HPV16 transgenic mouse model of squamous carcinogenesis, Terry R. Medler at Oregon Health & Science University in Portland, USA and his colleagues report that urokinase (uPA)+ macrophages regulate C3-independent release of C5a during premalignant progression, which in turn regulates protumorigenic properties of C5aR1+ mast cells and macrophages, including suppression of CD8+ T cell cytotoxicity. Therapeutic inhibition of C5aR1 via the peptide antagonist PMX-53 improved efficacy of paclitaxel chemotherapy associated with increased presence and cytotoxic properties of CXCR3+ effector memory CD8+ T cells in carcinomas, dependent on both macrophage transcriptional programming and IFNγ. Together, these data identify C5aR1-dependent signaling as an important immunomodulatory program in neoplastic tissue tractable for combinatorial cancer immunotherapy.



Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30417-3

2. Enhanced Anti-lymphoma Activity of CAR19-iNKT Cells Underpinned by Dual CD19 and CD1d Targeting

Chimeric antigen receptor anti-CD19 (CAR19)-T cell immunotherapy-induced clinical remissions in CD19+ B cell lymphomas are often short lived. Antonia Rotolo at Imperial College in London, UK and his colleagues tested whether CAR19-engineering of the CD1d-restricted invariant natural killer T (iNKT) cells would result in enhanced anti-lymphoma activity. CAR19-iNKT cells co-operatively activated by CD1d- and CAR19-CD19-dependent interactions are more effective than CAR19-T cells against CD1d-expressing lymphomas in vitro and in vivo. The swifter in vivo anti-lymphoma activity of CAR19-iNKT cells and their enhanced ability to eradicate brain lymphomas underpinned an improved tumor-free and overall survival. CD1D transcriptional de-repression by all-trans retinoic acid results in further enhanced cytotoxicity of CAR19-iNKT cells against CD19+ chronic lymphocytic leukemia cells. Thus, iNKT cells are a highly efficient platform for CAR-based immunotherapy of lymphomas and possibly other CD1d-expressing cancers.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30377-5

3. Aberrant ERBB4-SRC Signaling as a Hallmark of Group 4 Medulloblastoma Revealed by Integrative Phosphoproteomic Profiling

The current consensus recognizes four main medulloblastoma subgroups (wingless, Sonic hedgehog, group 3 and group 4). While medulloblastoma subgroups have been characterized extensively at the (epi-)genomic and transcriptomic levels, the proteome and phosphoproteome landscape remain to be comprehensively elucidated. Using quantitative (phospho)-proteomics in primary human medulloblastomas, Antoine Forget at Institut Curie, PSL Research University in Orsay, France and his colleagues unravel distinct posttranscriptional regulation leading to highly divergent oncogenic signaling and kinase activity profiles in groups 3 and 4 medulloblastomas. Specifically, proteomic and phosphoproteomic analyses identify aberrant ERBB4-SRC signaling in group 4. Hence, enforced expression of an activated SRC combined with p53 inactivation induces murine tumors that resemble group 4 medulloblastoma. Therefore, Their integrative proteogenomics approach unveils an oncogenic pathway and potential therapeutic vulnerability in the most common medulloblastoma subgroup.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30356-8

4. Proteomics, Post-translational Modifications, and Integrative Analyses Reveal Molecular Heterogeneity within Medulloblastoma Subgroups

There is a pressing need to identify therapeutic targets in tumors with low mutation rates such as the malignant pediatric brain tumor medulloblastoma. To address this challenge, Tenley C. Archer at Boston Children's Hospital in Boston, USA and his colleagues quantitatively profiled global proteomes and phospho-proteomes of 45 medulloblastoma samples. Integrated analyses revealed that tumors with similar RNA expression vary extensively at the post-transcriptional and post-translational levels. They identified distinct pathways associated with two subsets of SHH tumors, and found post-translational modifications of MYC that are associated with poor outcomes in group 3 tumors. They found kinases associated with subtypes and showed that inhibiting PRKDC sensitizes MYC-driven cells to radiation. Their study shows that proteomics enables a more comprehensive, functional readout, providing a foundation for future therapeutic strategies.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30358-1

5. The Oncogenic Transcription Factor RUNX1/ETO Corrupts Cell Cycle Regulation to Drive Leukemic Transformation

Oncogenic transcription factors such as the leukemic fusion protein RUNX1/ETO, which drives t(8;21) acute myeloid leukemia (AML), constitute cancer-specific but highly challenging therapeutic targets. Natalia Martinez-Soria at Newcastle University in Newcastle upon Tyne, UK and his colleagues used epigenomic profiling data for an RNAi screen to interrogate the transcriptional network maintaining t(8;21) AML. This strategy identified Cyclin D2 (CCND2) as a crucial transmitter of RUNX1/ETO-driven leukemic propagation. RUNX1/ETO cooperates with AP-1 to drive CCND2 expression. Knockdown or pharmacological inhibition of CCND2 by an approved drug significantly impairs leukemic expansion of patient-derived AML cells and engraftment in immunodeficient murine hosts. Their data demonstrate that RUNX1/ETO maintains leukemia by promoting cell cycle progression and identifies G1 CCND-CDK complexes as promising therapeutic targets for treatment of RUNX1/ETO-driven AML.

Read more, please click https://www.cell.com/cancer-cell/fulltext/S1535-6108(18)30375-1

Staining Proteins in Gels

Silver staining is used for sensitive detection of proteins separated by 1D and 2D SDS-PAGE with detection limits from 0.5-5 ng based on the selectively reduction of silver ions at sites of the gel that contain proteins and other macromolecules. Many silver staining protocols and commercial staining kits are not compatible with mass spectrometry due to the use of cross-linking reagents. Commercial silver staining kits compatible with mass spectrometry are available from different suppliers. Here we describe a silver staining protocol that has been optimized for mass spectrometric analysis. The protocol results in confdent protein identifcations and high sequence coverage by MALDI MS and ESI MS due to a high recovery of peptides from the stained gel. The protocol has been tested and documented in many publications (Mortz, E et al, Proteomics 1 (11), 1359-63, 2001).

Silver stained gel


Procedure:
1. Run 1D or 2D gel. Incubate the gel in Fixer (40% ethanol, 10% acetic acid, 50% H2O) for 1 hr.
2. Wash the gel in H2O for at least 30 min.
Note: Overnight washing with several changes of water will remove all acetic acid, reduce background staining and increase sensitivity.
3. Sensitize the gel in 0.02% sodium thiosulfate (0.04 g Na2S2O3, 200 ml H2O) for only 1 min.
Note: Longer time will decrease peptide recovery from the gel.
4. Wash gel in H2O for 3 x 20 sec.
5. Incubate gel for 20 min in 4ºC cold 0.1% silver nitrate solution (0.2 g AgNO3, 200 ml H2O, 0.02% formaldehyde, add 40μL 35% formaldehyde just before use).
Note: Staining is enhanced with cold AgNO3.
6. Wash the gel in H2O for 3 x 20 sec.
7. Place the gel in a new staining tray.
Note: Residual AgNO3 on the gel surface and staining tray will increase background staining.
8. Wash the gel in H2O for 1 min.
9. Develop the gel in 3% sodium carbonate (7.5 g Na2CO3 in 250 ml H2O), 0.05% formaldehyde (add 125 μL 35% formaldehyde just before use).
Note: Change developer solution immediately when it turns yellow. Terminate when the staining is suffcient.
10. Wash the gel in H2O for 20 sec.
11. Terminate staining in 5% acetic acid for 5 min.
12. Leave the gel at 4ºC in 1% acetic for storage. Prior to MS analysis the gel is washed in water for 3 x 10 min to ensure complete removal of acetic acid.

2018年10月17日星期三

Staining Proteins in Gels

Silver staining is used for sensitive detection of proteins separated by 1D and 2D SDS-PAGE with detection limits from 0.5-5 ng based on the selectively reduction of silver ions at sites of the gel that contain proteins and other macromolecules. Many silver staining protocols and commercial staining kits are not compatible with mass spectrometry due to the use of cross-linking reagents. Commercial silver staining kits compatible with mass spectrometry are available from different suppliers. Here we describe a silver staining protocol that has been optimized for mass spectrometric analysis. The protocol results in confdent protein identifcations and high sequence coverage by MALDI MS and ESI MS due to a high recovery of peptides from the stained gel. The protocol has been tested and documented in many publications (Mortz, E et al, Proteomics 1 (11), 1359-63, 2001).

Silver stained gel


Procedure:
1. Run 1D or 2D gel. Incubate the gel in Fixer (40% ethanol, 10% acetic acid, 50% H2O) for 1 hr.
2. Wash the gel in H2O for at least 30 min.
Note: Overnight washing with several changes of water will remove all acetic acid, reduce background staining and increase sensitivity.
3. Sensitize the gel in 0.02% sodium thiosulfate (0.04 g Na2S2O3, 200 ml H2O) for only 1 min.
Note: Longer time will decrease peptide recovery from the gel.
4. Wash gel in H2O for 3 x 20 sec.
5. Incubate gel for 20 min in 4ºC cold 0.1% silver nitrate solution (0.2 g AgNO3, 200 ml H2O, 0.02% formaldehyde, add 40μL 35% formaldehyde just before use).
Note: Staining is enhanced with cold AgNO3.
6. Wash the gel in H2O for 3 x 20 sec.
7. Place the gel in a new staining tray.
Note: Residual AgNO3 on the gel surface and staining tray will increase background staining.
8. Wash the gel in H2O for 1 min.
9. Develop the gel in 3% sodium carbonate (7.5 g Na2CO3 in 250 ml H2O), 0.05% formaldehyde (add 125 μL 35% formaldehyde just before use).
Note: Change developer solution immediately when it turns yellow. Terminate when the staining is suffcient.
10. Wash the gel in H2O for 20 sec.
11. Terminate staining in 5% acetic acid for 5 min.
12. Leave the gel at 4ºC in 1% acetic for storage. Prior to MS analysis the gel is washed in water for 3 x 10 min to ensure complete removal of acetic acid.

2018年10月15日星期一

The Nobel Prize in Physiology or Medicine 2001

The Nobel Prize in Physiology or Medicine 2001 was awarded jointly to Leland H. Hartwell, Tim Hunt and Sir Paul M. Nurse "for their discoveries of key regulators of the cell cycle."













NobelistBornAffiliation at the time of the award
Leland H. Hartwell30 October 1939, Los Angeles, CA, USAFred Hutchinson Cancer Research Center, Seattle, WA, USA
Tim Hunt19 February 1943, Neston, United KingdomImperial Cancer Research Fund, London, United Kingdom
Sir Paul M. Nurse25 January 1949, Norwich, United KingdomImperial Cancer Research Fund, London, United Kingdom
Summary

All organisms consist of cells that multiply through cell division. An adult human being has approximately 100 000 billion cells, all originating from a single cell, the fertilized egg cell. In adults there is also an enormous number of continuously dividing cells replacing those dying. Before a cell can divide it has to grow in size, duplicate its chromosomes and separate the chromosomes for exact distribution between the two daughter cells. These different processes are coordinated in the cell cycle.

This year’s Nobel Laureates in Physiology or Medicine have made seminal discoveries concerning the control of the cell cycle. They have identified key molecules that regulate the cell cycle in all eukaryotic organisms, including yeasts, plants, animals and human. These fundamental discoveries have a great impact on all aspects of cell growth. Defects in cell cycle control may lead to the type of chromosome alterations seen in cancer cells. This may in the long term open new possibilities for cancer treatment.



More details, please click The Nobel Prize in Physiology or Medicine 2001.

2018年10月14日星期日

The Nobel Prize in Physiology or Medicine 2001

The Nobel Prize in Physiology or Medicine 2001 was awarded jointly to Leland H. Hartwell, Tim Hunt and Sir Paul M. Nurse "for their discoveries of key regulators of the cell cycle."













NobelistBornAffiliation at the time of the award
Leland H. Hartwell30 October 1939, Los Angeles, CA, USAFred Hutchinson Cancer Research Center, Seattle, WA, USA
Tim Hunt19 February 1943, Neston, United KingdomImperial Cancer Research Fund, London, United Kingdom
Sir Paul M. Nurse25 January 1949, Norwich, United KingdomImperial Cancer Research Fund, London, United Kingdom
Summary

All organisms consist of cells that multiply through cell division. An adult human being has approximately 100 000 billion cells, all originating from a single cell, the fertilized egg cell. In adults there is also an enormous number of continuously dividing cells replacing those dying. Before a cell can divide it has to grow in size, duplicate its chromosomes and separate the chromosomes for exact distribution between the two daughter cells. These different processes are coordinated in the cell cycle.

This year’s Nobel Laureates in Physiology or Medicine have made seminal discoveries concerning the control of the cell cycle. They have identified key molecules that regulate the cell cycle in all eukaryotic organisms, including yeasts, plants, animals and human. These fundamental discoveries have a great impact on all aspects of cell growth. Defects in cell cycle control may lead to the type of chromosome alterations seen in cancer cells. This may in the long term open new possibilities for cancer treatment.



More details, please click The Nobel Prize in Physiology or Medicine 2001.

2018年10月11日星期四

A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes

Content introduction:

  • Nondestructive, base-resolution sequencing of 5-hydroxymethylcytosine using a DNA deaminase

  • De novo domestication of wild tomato using genome editing

  • A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes

  • Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A

  • Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells


1. Nondestructive, base-resolution sequencing of 5-hydroxymethylcytosine using a DNA deaminase

Here Emily K Schutsky at University of Pennsylvania in Philadelphia, Pennsylvania, USA and his colleagues present APOBEC-coupled epigenetic sequencing (ACE-seq), a bisulfite-free method for localizing 5-hydroxymethylcytosine (5hmC) at single-base resolution with low DNA input. The method builds on the observation that AID/APOBEC family DNA deaminase enzymes can potently discriminate between cytosine modification states and exploits the non-destructive nature of enzymatic, rather than chemical, deamination. ACE-seq yielded high-confidence 5hmC profiles with at least 1,000-fold less DNA input than conventional methods. Applying ACE-seq to generate a base-resolution map of 5hmC in tissue-derived cortical excitatory neurons, they found that 5hmC was almost entirely confined to CG dinucleotides. The whole-genome map permitted cytosine, 5-methylcytosine (5mC) and 5hmC to be parsed and revealed genomic features that diverged from global patterns, including enhancers and imprinting control regions with high and low 5hmC/5mC ratios, respectively. Enzymatic deamination overcomes many challenges posed by bisulfite-based methods, thus expanding the scope of epigenome profiling to include scarce samples and opening new lines of inquiry regarding the role of cytosine modifications in genome biology.

Read more, please click https://www.nature.com/articles/nbt.4204

2. De novo domestication of wild tomato using genome editing

Breeding of crops over millennia for yield and productivity has led to reduced genetic diversity. As a result, beneficial traits of wild species, such as disease resistance and stress tolerance, have been lost. We devised a CRISPR–Cas9 genome engineering strategy to combine agronomically desirable traits with useful traits present in wild lines. Agustin Zsögön at Universidade Federal de Viçosa in Viçosa, Brazil and his colleagues report that editing of six loci that are important for yield and productivity in present-day tomato crop lines enabled de novo domestication of wild Solanum pimpinellifolium. Engineered S. pimpinellifolium morphology was altered, together with the size, number and nutritional value of the fruits. Compared with the wild parent, their engineered lines have a threefold increase in fruit size and a tenfold increase in fruit number. Notably, fruit lycopene accumulation is improved by 500% compared with the widely cultivated S. lycopersicum. Their results pave the way for molecular breeding programs to exploit the genetic diversity present in wild plants.

Read more, please click https://www.nature.com/articles/nbt.4272

3. A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes

In the human malaria vector Anopheles gambiae, the gene doublesex (Agdsx) encodes two alternatively spliced transcripts, dsx-female (AgdsxF) and dsx-male (AgdsxM), that control differentiation of the two sexes. The female transcript, unlike the male, contains an exon (exon 5) whose sequence is highly conserved in all Anopheles mosquitoes so far analyzed. Kyros Kyrou at Imperial College in London, UK and his colleagues found that CRISPR–Cas9-targeted disruption of the intron 4–exon 5 boundary aimed at blocking the formation of functional AgdsxF did not affect male development or fertility, whereas females homozygous for the disrupted allele showed an intersex phenotype and complete sterility. A CRISPR–Cas9 gene drive construct targeting this same sequence spread rapidly in caged mosquitoes, reaching 100% prevalence within 7–11 generations while progressively reducing egg production to the point of total population collapse. Owing to functional constraint of the target sequence, no selection of alleles resistant to the gene drive occurred in these laboratory experiments. Cas9-resistant variants arose in each generation at the target site but did not block the spread of the drive.



Read more, please click https://www.nature.com/articles/nbt.4245

4. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A

Base editors (BEs) have been used to create C-to-T substitutions in various organisms. However, editing with rat APOBEC1-based BE3 is limited to a 5-nt sequence editing window and is inefficient in GC contexts. Here, Yuan Zong at Chinese Academy of Sciences in Beijing, China and his colleagues show that a base editor fusion protein composed of Cas9 nickase and human APOBEC3A (A3A-PBE) converts cytidine to thymidine efficiently in wheat, rice and potato with a 17-nucleotide editing window at all examined sites, independent of sequence context.

Read more, please click https://www.nature.com/articles/nbt.4261

5. Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells

Spatially resolved single-cell RNA sequencing (scRNAseq) is a powerful approach for inferring connections between a cell's identity and its position in a tissue. Keren Bahar Halpern at Weizmann Institute of Science in Rehovot, Israel and his colleagues recently combined scRNAseq with spatially mapped landmark genes to infer the expression zonation of hepatocytes. However, determining zonation of small cells with low mRNA content, or without highly expressed landmark genes, remains challenging. Here they used paired-cell sequencing, in which mRNA from pairs of attached mouse cells were sequenced and gene expression from one cell type was used to infer the pairs' tissue coordinates. They applied this method to pairs of hepatocytes and liver endothelial cells (LECs). Using the spatial information from hepatocytes, they reconstructed LEC zonation and extracted a landmark gene panel that they used to spatially map LEC scRNAseq data. Their approach revealed the expression of both Wnt ligands and the Dkk3 Wnt antagonist in distinct pericentral LEC sub-populations. This approach can be used to reconstruct spatial expression maps of non-parenchymal cells in other tissues.

Read more, please click https://www.nature.com/articles/nbt.4231

A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes

Content introduction:

  • Nondestructive, base-resolution sequencing of 5-hydroxymethylcytosine using a DNA deaminase

  • De novo domestication of wild tomato using genome editing

  • A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes

  • Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A

  • Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells


1. Nondestructive, base-resolution sequencing of 5-hydroxymethylcytosine using a DNA deaminase

Here Emily K Schutsky at University of Pennsylvania in Philadelphia, Pennsylvania, USA and his colleagues present APOBEC-coupled epigenetic sequencing (ACE-seq), a bisulfite-free method for localizing 5-hydroxymethylcytosine (5hmC) at single-base resolution with low DNA input. The method builds on the observation that AID/APOBEC family DNA deaminase enzymes can potently discriminate between cytosine modification states and exploits the non-destructive nature of enzymatic, rather than chemical, deamination. ACE-seq yielded high-confidence 5hmC profiles with at least 1,000-fold less DNA input than conventional methods. Applying ACE-seq to generate a base-resolution map of 5hmC in tissue-derived cortical excitatory neurons, they found that 5hmC was almost entirely confined to CG dinucleotides. The whole-genome map permitted cytosine, 5-methylcytosine (5mC) and 5hmC to be parsed and revealed genomic features that diverged from global patterns, including enhancers and imprinting control regions with high and low 5hmC/5mC ratios, respectively. Enzymatic deamination overcomes many challenges posed by bisulfite-based methods, thus expanding the scope of epigenome profiling to include scarce samples and opening new lines of inquiry regarding the role of cytosine modifications in genome biology.

Read more, please click https://www.nature.com/articles/nbt.4204

2. De novo domestication of wild tomato using genome editing

Breeding of crops over millennia for yield and productivity has led to reduced genetic diversity. As a result, beneficial traits of wild species, such as disease resistance and stress tolerance, have been lost. We devised a CRISPR–Cas9 genome engineering strategy to combine agronomically desirable traits with useful traits present in wild lines. Agustin Zsögön at Universidade Federal de Viçosa in Viçosa, Brazil and his colleagues report that editing of six loci that are important for yield and productivity in present-day tomato crop lines enabled de novo domestication of wild Solanum pimpinellifolium. Engineered S. pimpinellifolium morphology was altered, together with the size, number and nutritional value of the fruits. Compared with the wild parent, their engineered lines have a threefold increase in fruit size and a tenfold increase in fruit number. Notably, fruit lycopene accumulation is improved by 500% compared with the widely cultivated S. lycopersicum. Their results pave the way for molecular breeding programs to exploit the genetic diversity present in wild plants.

Read more, please click https://www.nature.com/articles/nbt.4272

3. A CRISPR–Cas9 gene drive targeting doublesex causes complete population suppression in caged Anopheles gambiae mosquitoes

In the human malaria vector Anopheles gambiae, the gene doublesex (Agdsx) encodes two alternatively spliced transcripts, dsx-female (AgdsxF) and dsx-male (AgdsxM), that control differentiation of the two sexes. The female transcript, unlike the male, contains an exon (exon 5) whose sequence is highly conserved in all Anopheles mosquitoes so far analyzed. Kyros Kyrou at Imperial College in London, UK and his colleagues found that CRISPR–Cas9-targeted disruption of the intron 4–exon 5 boundary aimed at blocking the formation of functional AgdsxF did not affect male development or fertility, whereas females homozygous for the disrupted allele showed an intersex phenotype and complete sterility. A CRISPR–Cas9 gene drive construct targeting this same sequence spread rapidly in caged mosquitoes, reaching 100% prevalence within 7–11 generations while progressively reducing egg production to the point of total population collapse. Owing to functional constraint of the target sequence, no selection of alleles resistant to the gene drive occurred in these laboratory experiments. Cas9-resistant variants arose in each generation at the target site but did not block the spread of the drive.



Read more, please click https://www.nature.com/articles/nbt.4245

4. Efficient C-to-T base editing in plants using a fusion of nCas9 and human APOBEC3A

Base editors (BEs) have been used to create C-to-T substitutions in various organisms. However, editing with rat APOBEC1-based BE3 is limited to a 5-nt sequence editing window and is inefficient in GC contexts. Here, Yuan Zong at Chinese Academy of Sciences in Beijing, China and his colleagues show that a base editor fusion protein composed of Cas9 nickase and human APOBEC3A (A3A-PBE) converts cytidine to thymidine efficiently in wheat, rice and potato with a 17-nucleotide editing window at all examined sites, independent of sequence context.

Read more, please click https://www.nature.com/articles/nbt.4261

5. Paired-cell sequencing enables spatial gene expression mapping of liver endothelial cells

Spatially resolved single-cell RNA sequencing (scRNAseq) is a powerful approach for inferring connections between a cell's identity and its position in a tissue. Keren Bahar Halpern at Weizmann Institute of Science in Rehovot, Israel and his colleagues recently combined scRNAseq with spatially mapped landmark genes to infer the expression zonation of hepatocytes. However, determining zonation of small cells with low mRNA content, or without highly expressed landmark genes, remains challenging. Here they used paired-cell sequencing, in which mRNA from pairs of attached mouse cells were sequenced and gene expression from one cell type was used to infer the pairs' tissue coordinates. They applied this method to pairs of hepatocytes and liver endothelial cells (LECs). Using the spatial information from hepatocytes, they reconstructed LEC zonation and extracted a landmark gene panel that they used to spatially map LEC scRNAseq data. Their approach revealed the expression of both Wnt ligands and the Dkk3 Wnt antagonist in distinct pericentral LEC sub-populations. This approach can be used to reconstruct spatial expression maps of non-parenchymal cells in other tissues.

Read more, please click https://www.nature.com/articles/nbt.4231

2018年10月8日星期一

15%-25% off on Abbkine top selling immunological and cytological products

Abbkine continually strive to provide innovative, high quality assay kits, recombinant proteins, antibodies and other research tools to accelerate life science fundamental research, drug discovery, etc. We focus on Immunology and Cytology.

Here, we choose some of our top selling products in immunology and cytology research to give you extra discount to help you with your research.

Promotion

Items: Products listed below.

Contents: Place order for 2 different items can enjoy 15% discount, more than 4 different items enjoy 20% discount.

Valid date: From 1st, October to 31th, December, 2018

Promotion List of Products

































































Product NameCatalog No.
ExKine™ Nuclear and Cytoplasmic Protein Extraction KitKTP3001
ExKine™ Nuclear Protein Extraction KitKTP3002
ExKine™ Cytoplasmic Protein Extraction KitKTP3003
ExKine™ Total Membrane Protein Extraction KitKTP3004
ExKine™ Membrane and Cytoplasmic Protein Extraction KitKTP3005
Protein Quantification Kit (BCA Assay)KTD3001
Protein Quantification Kit (Bradford Assay)KTD3002
Protein Gel Flash Staining KitK21010
SuperLumia ECL HRP Substrate KitK22020
SuperLumia ECL Plus HRP Substrate KitK22030
Colorcode Prestained Protein Marker (10-180 kDa)BMM3001
Colorcode Prestained Protein Marker (15-130 kDa)BMM3002
Cell Counting Kit-8 (CCK-8)KTC011001
Annexin V-AbFluor™ 405 Apoptosis Detection KitKTA0001
Annexin V-AbFluor™ 488 Apoptosis Detection KitKTA0002
Annexin V-AbFluor™ 555 Apoptosis Detection KitKTA0003
Annexin V-AbFluor™ 647 Apoptosis Detection KitKTA0004
Live and Dead Cell Double Staining KitKTA1001
Live Cell Tracking Kit (Orange Fluorescence)KTA1002
Mitochondrial Membrane Potential Assay Kit (JC-1)KTA4001
TraKine™ Cell Plasma Membrane Staining Kit (Green Fluorescence)KTC4001
TraKine™ Cell Plasma Membrane Staining Kit (Orange Fluorescence)  KTC4002
TraKine™ Mitochondrion Staining Kit (Green Fluorescence)KTC4003
TraKine™ Mitochondrion Staining Kit (Orange Fluorescence)KTC4004
TraKine™ Mitochondrion and Nuclear Staining Kit  KTC4005
TraKine™ F-actin Staining Kit (Green Fluorescence)KTC4008
TraKine™ F-actin Staining Kit (Orange Fluorescence)KTC4009
ExKine™ Nuclei Extraction KitKTP4001
ExKine™ Nuclei Extraction Kit (High Purity)KTP4002
ExKine™ Mitochondrion Extraction Kit (Cultured Cells)KTP4003
ExKine™ Mitochondrion Extraction Kit (Tissue)KTP4004
* This promotion is only available to end users. Abbkine reserves the right to cancel or refuse this promotion at any time.

15%-25% off on Abbkine top selling immunological and cytological products

Abbkine continually strive to provide innovative, high quality assay kits, recombinant proteins, antibodies and other research tools to accelerate life science fundamental research, drug discovery, etc. We focus on Immunology and Cytology.

Here, we choose some of our top selling products in immunology and cytology research to give you extra discount to help you with your research.

Promotion

Items: Products listed below.

Contents: Place order for 2 different items can enjoy 15% discount, more than 4 different items enjoy 20% discount.

Valid date: From 1st, October to 31th, December, 2018

Promotion List of Products

































































Product NameCatalog No.
ExKine™ Nuclear and Cytoplasmic Protein Extraction KitKTP3001
ExKine™ Nuclear Protein Extraction KitKTP3002
ExKine™ Cytoplasmic Protein Extraction KitKTP3003
ExKine™ Total Membrane Protein Extraction KitKTP3004
ExKine™ Membrane and Cytoplasmic Protein Extraction KitKTP3005
Protein Quantification Kit (BCA Assay)KTD3001
Protein Quantification Kit (Bradford Assay)KTD3002
Protein Gel Flash Staining KitK21010
SuperLumia ECL HRP Substrate KitK22020
SuperLumia ECL Plus HRP Substrate KitK22030
Colorcode Prestained Protein Marker (10-180 kDa)BMM3001
Colorcode Prestained Protein Marker (15-130 kDa)BMM3002
Cell Counting Kit-8 (CCK-8)KTC011001
Annexin V-AbFluor™ 405 Apoptosis Detection KitKTA0001
Annexin V-AbFluor™ 488 Apoptosis Detection KitKTA0002
Annexin V-AbFluor™ 555 Apoptosis Detection KitKTA0003
Annexin V-AbFluor™ 647 Apoptosis Detection KitKTA0004
Live and Dead Cell Double Staining KitKTA1001
Live Cell Tracking Kit (Orange Fluorescence)KTA1002
Mitochondrial Membrane Potential Assay Kit (JC-1)KTA4001
TraKine™ Cell Plasma Membrane Staining Kit (Green Fluorescence)KTC4001
TraKine™ Cell Plasma Membrane Staining Kit (Orange Fluorescence)  KTC4002
TraKine™ Mitochondrion Staining Kit (Green Fluorescence)KTC4003
TraKine™ Mitochondrion Staining Kit (Orange Fluorescence)KTC4004
TraKine™ Mitochondrion and Nuclear Staining Kit  KTC4005
TraKine™ F-actin Staining Kit (Green Fluorescence)KTC4008
TraKine™ F-actin Staining Kit (Orange Fluorescence)KTC4009
ExKine™ Nuclei Extraction KitKTP4001
ExKine™ Nuclei Extraction Kit (High Purity)KTP4002
ExKine™ Mitochondrion Extraction Kit (Cultured Cells)KTP4003
ExKine™ Mitochondrion Extraction Kit (Tissue)KTP4004
* This promotion is only available to end users. Abbkine reserves the right to cancel or refuse this promotion at any time.

15%-25% off on Abbkine top selling immunological and cytological products

Abbkine continually strive to provide innovative, high quality assay kits, recombinant proteins, antibodies and other research tools to accelerate life science fundamental research, drug discovery, etc. We focus on Immunology and Cytology.

Here, we choose some of our top selling products in immunology and cytology research to give you extra discount to help you with your research.

Promotion

Items: Products listed below.

Contents: Place order for 2 different items can enjoy 15% discount, more than 4 different items enjoy 20% discount.

Valid date: From 1st, October to 31th, December, 2018

Promotion List of Products

































































Product NameCatalog No.
ExKine™ Nuclear and Cytoplasmic Protein Extraction KitKTP3001
ExKine™ Nuclear Protein Extraction KitKTP3002
ExKine™ Cytoplasmic Protein Extraction KitKTP3003
ExKine™ Total Membrane Protein Extraction KitKTP3004
ExKine™ Membrane and Cytoplasmic Protein Extraction KitKTP3005
Protein Quantification Kit (BCA Assay)KTD3001
Protein Quantification Kit (Bradford Assay)KTD3002
Protein Gel Flash Staining KitK21010
SuperLumia ECL HRP Substrate KitK22020
SuperLumia ECL Plus HRP Substrate KitK22030
Colorcode Prestained Protein Marker (10-180 kDa)BMM3001
Colorcode Prestained Protein Marker (15-130 kDa)BMM3002
Cell Counting Kit-8 (CCK-8)KTC011001
Annexin V-AbFluor™ 405 Apoptosis Detection KitKTA0001
Annexin V-AbFluor™ 488 Apoptosis Detection KitKTA0002
Annexin V-AbFluor™ 555 Apoptosis Detection KitKTA0003
Annexin V-AbFluor™ 647 Apoptosis Detection KitKTA0004
Live and Dead Cell Double Staining KitKTA1001
Live Cell Tracking Kit (Orange Fluorescence)KTA1002
Mitochondrial Membrane Potential Assay Kit (JC-1)KTA4001
TraKine™ Cell Plasma Membrane Staining Kit (Green Fluorescence)KTC4001
TraKine™ Cell Plasma Membrane Staining Kit (Orange Fluorescence)  KTC4002
TraKine™ Mitochondrion Staining Kit (Green Fluorescence)KTC4003
TraKine™ Mitochondrion Staining Kit (Orange Fluorescence)KTC4004
TraKine™ Mitochondrion and Nuclear Staining Kit  KTC4005
TraKine™ F-actin Staining Kit (Green Fluorescence)KTC4008
TraKine™ F-actin Staining Kit (Orange Fluorescence)KTC4009
ExKine™ Nuclei Extraction KitKTP4001
ExKine™ Nuclei Extraction Kit (High Purity)KTP4002
ExKine™ Mitochondrion Extraction Kit (Cultured Cells)KTP4003
ExKine™ Mitochondrion Extraction Kit (Tissue)KTP4004
* This promotion is only available to end users. Abbkine reserves the right to cancel or refuse this promotion at any time.