文档库 最新最全的文档下载
当前位置:文档库 › 2004 CROH Cancer stem cells and differentiation therapy.

2004 CROH Cancer stem cells and differentiation therapy.

2004 CROH Cancer stem cells and differentiation therapy.
2004 CROH Cancer stem cells and differentiation therapy.

Critical Reviews in Oncology/Hematology51(2004)1–28

Stem cell origin of cancer and differentiation therapy

Stewart Sell?

Center and Ordway Research Institute,Empire State Plaza,New York State Health Department,Wadsworth Center,

P.O.Box509,Room C-400,Empire State Plaza,Albany,NY12201,USA

Accepted8April2004

Contents

Abstract (2)

1.Stem cells (2)

1.1.Embryonal,germinal and somatic(adult)stem cells (2)

1.2.Symmetric and asymmetric division (3)

1.3.Stem cell signals,development and cancer (4)

1.4.Potential/plasticity of stem cells (5)

1.5.Adult stem cells and normal tissue renewal (6)

1.6.Pleopotency of bone marrow stem cells (7)

2.Stem cells and cancer (7)

2.1.Embryonal rest hypothesis (7)

2.2.Anaplasia (8)

2.3.Maturation arrest (8)

3.Childhood tumors (8)

3.1.Wilms tumor (8)

3.2.Neuroblastomas (8)

4.Germinal stem cells and teratocarcinoma (9)

4.1.Totipotent embryonal cancer stem cells (9)

4.2.Teratocarcinomas (9)

4.3.Control of malignant potential of embryonal cancer cells (9)

4.4.Embryonic cells compared to embryonal carcinoma cells (10)

4.5.Regulation of tumor cell differentiation (10)

5.Skin cancer (10)

5.1.Skin cell lineage and malignant phenotype (10)

5.2.Skin cancers arise in self-renewing stem cells(initiation and promotion) (10)

5.3.Field cancerization (11)

6.Liver cancer (12)

6.1.Participation of various cells in the hepatocyte lineage in hepatocarcinogenesis (12)

6.2.Fusion of bone marrow and liver cells (13)

7.Adenocarcinomas (13)

7.1.Stem cell origin (13)

8.Hematopoietic stem cells and leukemia (13)

8.1.Normal blood renewal (13)

8.2.Multi/pluripotent hematopoietic stem cells (13)

8.3.Bone marrow transplantation and pleotypic potential (14)

8.4.Potentiality caveats (14)

8.5.Unanswered questions (15)

9.Leukemias as models of maturation arrest of a cell lineage (15)

9.1.Leukemia as a stem cell cancer (15)

9.2.Chemotherapy and reserve stem cells (15)

?Tel.:+15184740547;fax:+15184732900/4025381.

E-mail address:ssell@https://www.wendangku.net/doc/9f15720943.html,(S.Sell).

1040-8428/$–see front matter?2004Elsevier Ireland Ltd.All rights reserved.

doi:10.1016/j.critrevonc.2004.04.007

2S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–28

9.3.Gene translocations and leukemias (16)

9.3.1.The IL-3receptor (16)

9.3.2.B-cell lymphomas and the immunoglobulin heavy chain gene promoter (17)

9.4.Stage of maturation arrest and clinical course of leukemias (17)

10.Differentiation therapy (18)

10.1.Retinoic acids and teratocarcinomas (18)

10.2.Retinoids and acute promyelocytic leukemia(APL) (19)

10.3.Retinoid differentiation therapy;other cancers (20)

10.4.Chemoprevention of cancer by retinoids (20)

10.5.Retinoic acid syndrome (20)

11.Summary and conclusions (20)

Reviewers (21)

Acknowledgements (21)

References (21)

Biography (28)

Abstract

Our forefathers in pathology,on observing cancer tissue under the microscope in the mid-19th century,noticed the similarity between embryonic tissue and cancer,and suggested that tumors arise from embryo-like cells[1,2][Recherches dur le Traitement du Cancer,etc.Paris. (1829);Editoral Archiv fuer pathologische Anatomie und Physiologie und fuer klinische Medizin8(1855)23].The concept that adult tissues contain embryonic remnants that generally lie dormant,but that could be activated to become cancer was later formalized by Cohnheim [3,4][Path.Anat.Physiol.Klin.Med.40(1867)1–79;Virchows Arch.65(1875)64]and Durante[5][Arch.Memori ed Osservazioni di Chirugia Practica11(1874)217–226],as the“embryonal rest”theory of cancer.An updated version of the embryonal rest theory of cancer is that cancers arise from tissue stem cells in adults.Analysis of the cellular origin of carcinomas of different organs indicates that there is,in each instance,a determined stem cell required for normal tissue renewal that is the most likely cell of origin of carcinomas[6] [Lab.Investig.70(1994)6–22].In the present review,the nature of normal stem cells(embryonal,germinal and somatic)is presented and their relationships to cancer are further expanded.Cell signaling pathways shared by embryonic cells and cancer cells suggest a possible link between embryonic cells and cancer cells.Wilm’s tumors(nephroblastomas)and neuroblastomas are presented as possible tumors of embryonic rests in children.Teratocarcinoma is used as the classic example of the totipotent cancer stem cell which can be in?uenced by its environment to differentiate into a mature adult cell.The observation that“promotion”of an epidermal cancer may be accomplished months or even years after the initial exposure to carcinogen(“initiation”),implies that the original carcinogenic event occurs in a long-lived epithelial stem cell population.The cellular events during hepatocarcinogenesis illustrate that cancers may arise from cells at various stages of differentiation in the hepatocyte lineage.Examples of genetic mutations in epithelial and hematopoietic cancers show how speci?c alterations in gene expression may be manifested as maturation arrest of a cell lineage at a speci?c stage of differentation.Understanding the signals that control normal development may eventually lead us to insights in treating cancer by inducing its differentiation(differentiation therapy). Retinoid acid(RA)induced differentiation therapy has acquired a therapeutic niche in treatment of acute promyelocytic leukemia and the ability of RA to prevent cancer is currently under examination.

?2004Elsevier Ireland Ltd.All rights reserved.

Keywords:Stem cells;Cancer;Differentiation;Plasticity;Carcinogenesis;Embryonal rests;Differentiation therapy;Retinoic acid

“The simplest view appears to me undoubtedly to be that in an early stage of embryonic development more cells are produced than are required for building up the part concerned,so that there remains unappropriated a quantity of cells--it may be very few in number--which,owing to their embryonic character,are endowed with a marked capacity for proliferation...The only point on which I lay stress is that the real cause of the subsequent tumour is to be sought in a fault or irregularity of the embryonic rudiment.”Julius Cohnheim,1889

“Oncogeny is blocked ontogeny.”Van R.Potter,1968“Cancer is a problem of developmental biology.”G.Barry Pierce et al.19781.Stem cells

1.1.Embryonal,germinal and somatic(adult)stem cells There are three kinds of stem cells:embryonal,germinal, and somatic or adult stem cells[7].Embryonal stem cells are derived from the?rst?ve or six divisions of the fertilized egg.The progeny of embryonal stem cells are the precursors for all of the cells of the adult organs.Germinal stem cells in the adult produce eggs and sperm and are responsible for reproduction.Somatic stem or progenitor cells are consid-ered more limited in their potential,and they produce cells that differentiate into mature functioning cells and that are responsible for normal tissue renewal.The ultimate stem cell,the fertilized egg and its embryonic stem cell progeny

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–28

3

Fig.1.Stem cell lineages in adult tissues and normal tissue renewal.In analogy to the structure of a tree with a very small trunk,normal tissue stem cells represent a very small proportion of the tissue cells and do not normally proliferate.The differentiating progeny of the tissue-determined stem cells is represented by the branches,which represent lineages of cells eventually giving rise to the terminally differentiated functional parenchymal cells, represented by the leaves.For deciduous trees,loss of the leaves at the end of the year(falling off or apoptosis)occurs in cycles,whereas in animal tissues there is a continuous production and loss of cells.Cancer results when the rate of proliferation increases and the rate of cell loss remains the same or when the rate of proliferation stays the same and/or the rate of loss decreases.

are totipotent and give rise to branches(lines)of cells that form various differentiated organs.During this process,the progeny of the embryonic stem cells lose potential and gain differentiated properties,in a poorly understood process called determination.In the adult,the cells responsible for tissue renewal are no longer totipotent,but become more re-stricted in their ability to form different tissues.An exception to this rule may be germinal cells.Some tumors of germinal cells(embryonal carcinomas)are able to produce differenti-ated cell types of all adult organs,as well as placental tissues (see below).It is generally accepted that adult tissues contain tissue-determined stem cells responsible for normal tissue renewal.However,there is increasing evidence for retention of some stem cells in the non-germinal tissues of adults,es-pecially in the bone marrow,that retain the potential to pro-duce different cell lineages.These“reserve”stem cells are very few in number as most of the cellular renewal is accom-plished by tissue determined transit-amplifying cells(Fig.1).

1.2.Symmetric and asymmetric division

The various types of stem cells not only have different potentials,but they also proliferate differently.Cells can di-vide symmetrically,whereby each daughter cell retains the properties of the parental cells,or asymmetrically,whereby one daughter cell retains the properties of the parental stem cell,whereas the other daughter cell begins the process of determination(Fig.2)[8].The characteristic of embryonal stem cells is that they divide symmetrically.Each daughter cell remains a totipotent stem cell,resulting in a logarithmic expansion of cells during early embryonic growth.Then,as the germ layers of the early embryo form and the process of determination begins,the cells proliferate asymmetrically. The tissue determined stem(progenitor cells)divide asym-metrically as one daughter cell remains to continue the pro-cess of cell renewal and the other daughter cell starts the process of differentiation(transit amplifying cells).The de-termined transit amplifying cells retain the ability to divide for several differentiation stages and are the major contribu-tors to normal tissue renewal.These cells are known as pro-genitor cells in the bone marrow,and as transit-amplifying cells in the skin and other tissues.In these and other rapidly proliferating tissues the stem cells are normally quiescent and do not divide.The tissue is renewed by proliferation of the daughter cells(transit amplifying cells),which con-tinue the process of determination.Finally,the end product is a terminally differentiated cell(red blood cell or poly-morphonuclear cell in the blood;keratinized epithelial cell in the skin).

The molecular mechanism of asynchronous division is poorly understood.In1975,Cairns hypothesized that the reserve tissue stem cell retains an immortal reserve DNA template which is passed on each division to the reserve stem cells,whereas the transit amplifying cells receive only newly synthesized DNA[9].Although this is dif?cult to prove,there are recent data that this may occur in a tis-sue culture of a mouse?broblast cell line[10],as well as in the crypts of the small intestine[11].How this re-

4S.Sell /Critical Reviews in Oncology/Hematology 51(2004)

1–28

Fig.2.Symmetric and asymmetric division.When cells divide symmetrically,each daughter cell is identical and retains the potential of the parental cell.When cells divide asymmetrically,one of the daughter cells remains a stem cell,whereas the other begins the process of determination [310,311].In adult tissues,the progeny of the tissue stem cells undergoing differentiation (determination)retain the capacity to proliferate (transit-amplifying cells)and comprise the major source of cells for normal tissue renewal.

serve DNA functions to maintain the tissue stem cell is not known.

Asynchronous division may be determined by anatomical boundries.In germinal cells,one pole of an asymmetrically dividing cell contains different protein complexes than are present at the other pole [12,13]or one of the poles may be anchored to a supporting cell [14]or membrane by a cad-herin,?-catenin,APC (adenomatous polyposis coli)com-plex (see below),so that one daughter cell remains attached to a surrounding cell whereas the other does not and begins the process of determination [15].It appears that asymmet-ric division of adult stem cells can be suppressed by the purine nucleoside xanthosine,which stimulates guanine ri-bonucleotide synthesis and promotes symmetric clonal ex-pansion.This is reported to be a mechanism that can be used to derive liver stem cell lines from adult tissues [16].1.3.Stem cell signals,development and cancer

Understanding what controls the maintenance of stem cells and differentiation signals may give insights into the cellular signals involved in cancer,and may ultimately lead to new approaches to differentiation therapy.The signal that controls which daughter cell of an adult stem cell remains a stem cell and which begins the process of determination may be mediated through a number of signaling pathways including the Oct-4,Wnt/?-catenin,Notch,BMP (bone mor-phogenic protein),Janus family kinase,or sonic hedgehog signaling pathways,etc.[13,17–19].Two of these,Oct-4and the Wnt/?-catenin are presented as examples.

Oct-4is a transcription factor that is expressed only in the inner cell mass of the embryo,but not in the trophecto-derm,the structure that will form the extra-embryonic tis-sues.During later development Oct-4expression is restricted to cells of the germ line [20],and is essential in maintaining totipotency and synchronous division.Loss of expression is associated with differentiation of cells.The human papillo-mavirus E7oncoprotein speci?cally binds to a domain on the Oct-4promoter to activate Oct-4expression leading to viral transformation [21].Oct-4is a marker for embryonal cancer [22],and treatment with retinoids,which induce dif-ferentiation of embryonal carcinomas,causes a decrease in expression of Oct-4[23].Thus,Oct-4expression appears to be important in maintaining the undifferentiated state of embryonal carcinoma [24],as well as in other cancers.Un-derstanding how to block Oct-4promises to be an approach to controlling the differentiation state of some of the least differentiated cancers (differentiation therapy,see below).Other pathways are important at later stages of differenti-ation and activation of the genes controlling these pathways could act later in adult tissues as oncogenes [19],hold-ing cells in a state of maturation arrest.For example,the Wnt/?-catenin cell activation pathway (Fig.3)is active in maintaining proliferation at an early stage of differentiation and may have a similar role in cancer cells [18,25].Wnt signaling affects the orientation of the chromosomes during mitotic division,and abnormalities in the orientation might contribute to mitotic disjunctions typical of cancer cells [26].In general,Wnt/?-catenin signaling activates proliferation [27]and inhibits apoptosis [28,29],the classic hallmarks of cancer cells.However,this pathway may also be instru-mental for differentiation signals critical in determining the fate of cells during development of many different organs [17,30–34].Cell proliferation and differentiation in the

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–285

Fig.3.The Wnt/?-catenin signaling pathway.When this signaling path-way is not activated,?-catenin is bound to adherins in the junctions between cells,and unbound?-catenin is tagged for destruction by a complex of proteins including the adenomatous polyposis coli protein (APC).Activation of the Wnt signaling pathway by binding of Wnt to the seven-pass transmembrane Frizzled receptor leads to phosphorylation of the disheveled protein(Dsh),which then associates with axin and pre-vents glycogen synthase kinase3(Gsk-3)from phosphorylating critical substrates,the negative regulators axin and APC,as well as?-catenin itself[26].The unphosphorylated cellular?-catenin escapes recognition by?-TRCP,a component of an E2ubiquitin ligase,and is protected from ubiquinization and destruction.The cellular?-catenin then translocates to the nucleus,where it associates with transcription factors,such as Tcf and Lcf[43,312],and activates transcription of cell proliferation genes,in particular,cyclin D1,c-jun and c-myc[41,51].Modi?ed from[26,313]. wing of the?y Drosophila is controlled by region-speci?c signals that act through the Wnt pathway[35].Long-term hematopoietic stem cells show up-regulation of the frizzled Wnt receptor[36].Thus,Wnt/?-catenin signaling appears to be at least one of the critical means by which prolif-eration and differentiation of cells are controlled during development,and it could be predicted that this pathway also plays a critical role in cancer.

Wnt was recognized as a putative oncogene over20 years ago,before it was found to be the homologue of the Drosophila wingless(wnt-1)gene[37].Transfection of tis-sue culture cells,with the proto-oncogene c-wnt,results in accumulation of cellular?-catenin and morphologic trans-formation[38,39].Expression of a truncated,constitutively active form of?-catenin in mouse mammary gland causes multiple aggressive mammary carcinomas[40].In colon and other cancers,mutations in the adenomatous polypo-sis coli(APC)tumor suppressor protein or in?-catenin itself,stabilize?-catenin,enhancing its ability to activate transcription of cyclin D1[41]and to increase proliferation [42,43].In most colorectal cancers,mutations in APC result in an absence or loss of function of the protein,so that the phosphorylation of?-catenin by the GSK-3/axin complex is inhibited and?-catenin is not catabolized[44].A high concentration of cytoplasmic?-catenin results in continued growth of colorectal cancer cells[45].In addition,Wnt-2 is up-regulated in gastrointestinal cancer and detection of the Wnt-2protein in the feces may be used as a marker for cancer[46].Mutations in the?-catenin gene and overex-pression of?-catenin are found in most hepatoblastomas [47],and in about50%of human hepatocellular carcinomas (HCC)[27,48,49],as well as in HCC induced by chemicals in mice[50,51].Expression of?-catenin is associated with increased levels of cyclin D1,jun,and myc[51].

It is possible that treatment of cancers with increased ?-catenin as a molecular lesion may be accomplished by agents which cause a decrease in?-catenin.Exisulind and other inhibitors of cyclic GMP phosphodiesterases(sulin-dac sul?de,CP248,CP461)stimulate phosphorylation and degradation of?-catenin,both by proteosome degradation (protein kinase G mediated)and by capase activation,which is independent of GSK-3?,leading to apoptosis of cancer cells[52,53].Since the activities of exisulind and its ana-logues are not only independent of APC and GSK3?,but also of cyclooxygenase expression,p53mutations or bcl-2 expression[54],they may provide effective reversal of the molecular defects of the wnt-signalling pathway in cancers. In early clinical trials,exisulind has tolerable toxicity and shows some promise in combination with other drugs for chemotherapy of metastatic breast cancer[55].In the fu-ture other approaches for therapeutic intervention may be directed to the?-catenin pathway or to other cell signaling pathways.

1.4.Potential/plasticity of stem cells

The ability of a stem or progenitor cell to produce progeny that can express various mature phenotypes is called poten-tial or plasticity.The term totipotent should be reserved for those stem cells that can give rise to all of the differentiated tissues of the body as well as the placenta and membranes (embryonic stem cells;germinal stem cells).The terms mul-tipotent and pluripotent are essentially synonymous:both refer to the ability of a given stem cell to form many differ-ent cell types.In an of?cial NIH primer released in2000, pluripotent is de?ned as“capable of giving rise to most tissues of an organism”,and totipotent as“having unlimited capacity”(https://www.wendangku.net/doc/9f15720943.html,/news/stemcell/primer.htm).During the process of development,the totipotent embryonal stem cells give rise to multi/pluripotent progenitor cells of the germ layers.These,in turn,give rise to oligopotent pro-genitor cells of the developing organs(See Table1).The inner cell-mass cells of the embryo are multi/pluripotent [56].The bone marrow(hematopoietic)stem cells are also multi/pluripotent,and can normally produce red blood cells,platelets,polymorphonuclear leukocytes,monocytes and lymphocyte precursors[57].Most,but not all,of these become terminally differentiated in their fully ma-ture forms.The stromal cells of the bone marrow are also multi/pluripotent and can produce progeny that become

6

S.Sell /Critical Reviews in Oncology/Hematology 51(2004)1–28

Table 1

Terminology of potential (plasticity)Pre?x Meaning Example

Toti-All

Embryonal

Pluri-Several/many Inner cell mass/hematopoietic Multi-Many/much Inner cell mass/hematopoietic Pleo-a Excessive/more Hematopoietic,skin Oligo-Few/little GI stem cell Quadri-Four GI stem cell Tri-Three Bronchial lining Bi-Two Bile duct Uni-One

Prostate

Source:The Random House Dictionary of the English Language.

a Recent evidence indicates that some cells in the tissues used as examples of determined potential may have more potential than previously appreciated (pleopotential).For example,hematopoietic stem cells may give rise to many different mature cell types;skin stem cells,believed to be uni-or bi-potent,may also contain multipotent progenitor cells [63],and thus exhibit pleopotency.

osteoblasts,osteoclasts,chrondrocytes,?broblasts,or mus-cle cells.The intestinal progenitor is quadripotent;it can give rise to progeny that become mucous,absorptive,neu-roendocrine,or Paneth cells.The bronchial lining cells are tripotent:progeny become neuroendocrine,mucous or cili-ated cells.The bile ductular progenitor cells of the liver are bipotent;they produce duct cells and hepatocytes.

Under pathologic conditions,some of these progenitor cells appear to be able to “transdifferentiate”to a differ-ent cell type.For example,bronchial epithelial progenitor cells can produce progeny that become squamous epithe-lium.Hematopoietic and/or bone marrow stromal stem cells may be able to produce progeny that can differentiate into various different tissues including epithelial cells [57].Be-EMBRYONAL

STEM CELL

Fig.4.Relationship between stage of differentiation of cell lineage and cancer type.A hypothetical cell lineage,from totipotent stem cell to terminally differentiated cell,is depicted.It is hypothesized that a malignant event may be manifested at any point along the differentiation pathway.The degree of differentiation of the resultant cancer will be determined by the stage in the process of determination at which the malignant event occurs or is manifested.The concept of tissue stem cells giving rise to cancer represents a modern day version of the “embryonal rest”theory of cancer.Speci?c examples of this model for teratocarcinomas,skin and liver cancer,and hematogenous malignancies are presented in the text.Modi?ed form Pierce et al.[73].

cause the potential of these stem cells is less than totipotent,but more than the expected multi/pluripotency of the hep-atopoietic lineage,the term “pleopotent ”is suggested (see below).

1.5.Adult stem cells and normal tissue renewal

Normal tissue renewal is accomplished by tissue stem cells that divide to give rise to one daughter cell that re-mains a stem cell and another daughter cell that begins the process of determination (asynchronous division).In most organs,the normal replacement of terminally differentiated cells is accomplished by proliferation of progenitor cells or transit-amplifying cells.Transit-amplifying cells provide an expanded population of proliferating tissue determined cells and produce progeny that differentiate into more mature cells that can no longer proliferate and eventually die (see Fig.4).In classic embryology,determination is considered a unidirectional pathway [7].For example,when primitive endoderm becomes committed to forming liver,it is not able to dedifferentiate or to transdifferentiate into another tissue type.Classically,the term transdifferention was used for the process of reprogramming cells that have already embarked on a differentiation pathway,and has been used interchange-ably with metaplasia and transdetermination [58].However,it is now appreciated that adult progenitor cells of one tis-sue may be able to differentiate into mature cells of another tissue type.For example,the adult bone marrow contains small numbers of undifferentiated stem cells having suf?-cient plasticity to give rise to progeny that can become at least nine different mature cell types [19],depending on the environment in which they are seeded [57,59],a concept originally proposed by Cohnheim in 1867[3].According to

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–287

1. TISSUE DETERMINED ADULT STEM CELLS

2. PLEO/PLURIPOTENT ADULT STEM CELL

3. TRANSDIFFERENTIATON

4. DEDIFFERENTATION

5. FUSION

Fig.5.Five possible pathways for generation of different tissue types in adult tissues.1.From tissue determined stem cells.In the classic model of tissue determination,progenitor cells become committed(determined)for a speci?c tissue type.They then give rise to progeny that differentiate into the speci?c tissue.2.Pleo/pluripotent tissue stem cells.In tissues such as the blood,the tissue stem cell retains the potential to give rise to progeny that can differentiate into different cell lineages.3.Transdifferentiation of determined cells.Progenitor cells that have become determined for one tissue type change their determination and give rise to a different cell type.4.Dedifferentiation.Mature tissue cells,which still have the capacity to divide,undergo backwards differentiation and give rise to progeny that can differentiate into a different cell type.This has been demonstrated during amphibian limb regeneration[314].5.Fusion.It is possible that stem cells from one tissue(e.g.,bone marrow)may fuse with cells of another tissue(liver)to form “hybrid”cells that may re?ect new differentiation properties[164,165].

the original meaning of the term,this is not transdifferentia-

tion,rather,it is unexpected plasticity or“pleopotency”(see

below).Of the possible pathways for differentiation of adult

tissues shown in Fig.5,the most likely paths are1and2,

but not3,4,or5.However,the term transdifferentiation is

frequently used,or,in fact misued,when pleopotency would

be more appropriate.

1.6.Pleopotency of bone marrow stem cells

In1867,Cohnheim hypothesized that the blood contained

cells that can participate in normal tissue renewal of other

organs[3].In fact,it appears from accumulating evidence

that,not only does the bone marrow contain a blood-forming

stem cell,but also it contains an even more primitive cell

with greater potential.This more primitive stem cell pro-

duces not only cells of the hematopoietic lineage,but also

mesenchymal progenitor cells that can give rise to many

other cell types,such as osteocytes,adipocytes,muscle cells,

astrocytes,and neurons,as well as stromal cells that sup-

port hematopoiesis[57,59,60,61].In addition,it has the ca-

pacity to circulate to other organs and to replace different

non-hematopoietic tissues[57,59].Because this cell most

likely is not totipotent like embryonal stem cells,but never-

theless has unexpected potential,the term pleopotent(from

the Greek,excessive,more)is hereby suggested to describe

this cell.Ii is this cell in the adult that offers new approaches

to replacement of damaged tissue not previously considered

possible[62].

The true tissue stem cell normally divides very rarely,but

it can be stimulated to proliferate if there is a loss of,or

an increasing demand on,transit amplifying cells.This is

termed a reserve stem cell.For example,proliferation of the

resting bone marrow stem cells may be activated by blood

loss or by a decrease in the oxygen supply at high altitudes.

To maintain the total number of cells in an adult organ in

equilibrium,the number of progenitor cells that divide must

essentially equal the number of cells that differentiate and

die.If more cells are produced than die,either because the

number of proliferating cells is greater than the number of

cells dying,or because the number of dying cells is smaller

than the number of proliferating cells,then the number of

cells will increase,the primary feature of cancer.Under con-

ditions of temporary stress,such as high altitude oxygen re-

duction or blood loss,this increase is reversible;in cancer

it is not.

2.Stem cells and cancer

2.1.Embryonal rest hypothesis

The histologic resemblance of the tissue of teratocarci-

nomas to that of the developing fetus suggested to Virchow

in the mid-1800s[2,64]a possible relationship between the

two tissues.When compared to normal tissue,cancers were

termed dedifferentiated to denote the less-differentiated cel-

lular structure similar to fetal tissue.Today,pathologists

still refer to the“degree of dedifferentiation”,in the grading

of tumors:poorly-differentiated suggests a bad prognosis;

well-differentiated,a better prognosis.However,it is un-

fortunate that this term implies a process whereby tumors

8S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–28

arise from mature cells becoming immature cells-an unlikely event.Cohnheim[3,4,65]and Durante[5]postulated that cancer in adults develops from embryonal rudiments that are produced in excess and that remain in the tissues of the fully mature organs.In1867,Cohnheim[3]hypothesized that all tissues are renewed by cells in the blood stream;from this concept it follows that both normal tissues and cancer can arise from stem cells in the blood.Beard extended the embryonal rest hypothesis by concluding that tumors arise from displaced placental tissue or activated germinal cells in adult tissues(see Oberling[66]).

In1889,Cohnheim[65]further hypothesized that the be-nign or malignant character of a new growth depends solely on the behavior of the remainder of the organism.He stressed the importance of a blood supply,or angiogenesis,for pro-gression of cells from benign to malignant.Ribbert[67]in 1904proposed that the critical factor for expression of the malignant phenotype of these cells is their isolation from a normal controlling environment,a concept that will be ex-panded in consideration of teratocarcinomas below.Rotter [68]thought that primitive sex cells(germinal cells)wander through the tissue of the developing embryo and can acci-dentally lodge in tissues outside the ultimate sex glands,and can in these new locations serve as the origin of tumors. According to these concepts,all cancers might arise from embryonal-like progenitor cells or germinal cells present in the wrong places in adult tissues.Malignant cells and nor-mal stem cells share the ability to invade,migrate and grow in tissue sites distant from where they arose[69,70].We pos-tulate that tissue stem cells are the modern day equivalent of embryonal rests and that most tumors arise from matura-tion arrest of a cellular lineage derived from a tissue stem cell[6].

2.2.Anaplasia

The term“anaplasia”(ana,backward;plasis,to mold) was?rst coined to describe the change in cells that allows cancerous growth by dedifferentiation.The embryonal rest origin of cancer was generally replaced by the idea that external agents such as chemicals or viruses act on differen-tiated tissue cells to cause cancer,by inducing dedifferenti-ation of mature adult cells.Anaplasia is now used as a mor-phological term to describe a loss of differentiated features (without form),rather than as a term to describe a process. In modern cancer biology,genetic changes leading to alter-ations of growth regulatory functions are widely accepted as causing the altered growth seen in cancers[71].Chemical carcinogens and oncogenic viruses that cause these genetic changes must act on dividing progenitor cells,rather than on mature cells.An exception is the liver,an organ in which even the most mature cells retain the capacity to divide(see below).Thus,even if a carcinogen acts on a mature liver cell,this cell must proliferate in order for the carcinogenic event to be manifested.From a mathematical analysis of the total number of mutation events during the history of a cell,it is evident that mutations are likely to accumulate rapidly in cells during their symmetric linear growth phase (i.e.,during early development[72].It is hypothesized that this process results in the seeding of the tissues of a young individual with a small fraction of mutated stem cells that serve as the origin of cancer in the mature adult.

2.3.Maturation arrest

Cancers at any age may arise from the proliferating progeny of the stem cells(transit amplifying cells);the de-gree of differentiation depends on the stage of differentiation at which the oncogenic lesion is manifested(Fig.4;[6,73]). If maturation arrest occurs early in the determination of a cell lineage,the cancers will be poorly differentiated;if the arrest occurs later,they will be well differentiated.

In order for an alteration in a cell to cause cancer the change must arise in a cell that has the potential to divide and not be lost during normal tissue turnover.For example, the cells above the basal layer of the skin or in the upper mucosa of the gastrointestinal tract that undergo a malignant mutation are most likely sloughed off before they can give rise to a tumor mass.In order for a cancer to be produced, there must be a way for the transformed cells to remain in the body.This is the property of the tissue stem cells or the least-differentiated tissue determined stem cell,which can generate one daughter cell that remains in the tissue.

3.Childhood tumors

3.1.Wilms tumor

Tumors arising in children provide clues to the relation-ship of“residual”embryonic cells and cancer[2].In1899, Wilms[74]described a tumor of the kidney of children un-der8years of age,which contained a mixture of embry-onal cell types.He concluded that it arose from a fragment of the primitive undifferentiated mesodermal tissue,which normally is the anlage of the myotome(striated muscle), nephrotome(Wolf?an body),and smooth muscle.It is com-posed of a mixture of undifferentiated spindle cells,imma-ture epithelial tubules,and“rosettes”of cells similar to em-bryonal glomeruli(James Homer Wright rosettes),as well as sarcomatous tumor cells and nonstriated muscle,leading to the term“nephroblastoma”[75,76].Wilms’tumors ap-pear to arise from embryonal cells in infants;these embry-onic rests differentiate with age and essentially disappear, so that they are no longer a source of cancers after7years of age[76,77].

3.2.Neuroblastomas

Neuroblastomas are tumors of infants composed of a mix-ture undifferentiated spindle cells and immature epithelial tubules[78].Neuroblastomas arise in fetal neural crest cells

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–289

of the sympathetic nervous system.In young infants,they are anaplastic and aggressive,whereas in older children they appear as more differentiated tumors or even as benign ganglioneuromas[79].The undifferentiated embryonal-type neural tumor cells become more differentiated with age,and can differentiate into benign mature cells in the environment of the more mature child.The most likely instance of a tumor arising from stem cells is germinal cell teratocarcinoma. 4.Germinal stem cells and teratocarcinoma

4.1.Totipotent embryonal cancer stem cells

Tumors of germinal stem cells in the adult clearly demonstrate the totipotentiality and tumorigenicity of tu-mor stem cells.Teratocarcinomas of mice can be produced by transplantation of germinal cells from the genital ridges of21-day-old fetal mice into the testes of adult syngeneic mice[80,81],and have been maintained for hundreds of generations by serial transplantation or cultivation in vitro. Through a change in the tissue environment,the normal ger-minal stem cells acquire malignant growth potential.Gard-ner concluded that extrinsic factors in the environment are required to maintain normal differentiation of the germinal cells[58]–a concept avidly espoused earlier by Cohnheim, [65].When removed from this controlling environment,the otherwise normal germinal cells do not mature normally.

4.2.Teratocarcinomas

Teratocarcinomas are made up of many differentiated cell types representing essentially any normal cell of adult tis-sues including germinal cells as well as embryonic and fetal (yolk sac and placental)tissues[82].The malignant cells are the embryonic(embryonal carcinoma)cells[83–85], which form structures resembling pre-somite embryos,so called embryoid bodies(les boutons embryonnaires;[86]). The malignant core cells of teratocarcinomas cultured in vitro also form organized structures that resemble the early developing embryo.The cells that make up the embryoid body are undifferentiated,but have the capacity to differen-tiate into mature benign cells[87,88].Approximately10% of single embryonal carcinoma cells from the teratocarci-noma develop into tumors that can contain more than two dozen well-differentiated adult tissues,including brain,mus-cle,bone,teeth,bone marrow,eyes,secretory glands,skin and intestine,arising from all three germ layers as well as placenta and yolk sac[85].

In humans,teratocarcinomas appear mainly in young adults at any site along the migration pathway of germinal cells from the brain to the gonads during development[82]. The differentiation of the malignant undifferentiated embry-onal carcinoma cells results in an appearance of the tumor that is a caricature of normal tissue development[87,88]. The observation that cancers morphologically resemble the tissues from which they derive was emphasized by Cohn-heim[65](p.751)who wrote:“It is then self-evident that the elements of a tumor cell will closely follow the speci?c type of individual in whose body it arises,for example, in the tumours of man and mammals hairs and not feath-ers will be met with,and in the corresponding tumours of birds,always feathers.”However,the character of the cell of origin of teratocarcinomas,and not the tissue site of origin,determines the cellular makeup of the tumor,but the environment determines the malignant phenotype.The com-position of the mature cells in teratocarcinomas does not differ substantially at the different sites,with the possible exception that malignant ovarian teratocarcinomas appear to contain large amounts of neural tissue[89].Thus,it seems likely that teratocarcinomas arise from an undifferentiated germinal stem cell present in various tissues,usually along the developmental migration pathway of germinal cells. 4.3.Control of malignant potential of embryonal

cancer cells

The growth of teratocarcinomas appears to be due to loss of the growth restrictions that are placed on the developing germinal cells by their normal environment.The ability of the environment to control differentiation of the cancerous cells of teratocarcinomas was unequivocally demonstrated when transplantable teratocarcinoma cells were injected into the inner cell mass of a normal blastocyst[56,90–92].Not only is the blastocyst able to control the differentiation of these tumor cells,but the tumor stem cells are also incorpo-rated into the developing embryo,resulting in development of mosaic adult mice,with normally functioning tissues de-rived from the tumor cells[91].As determined by histocom-patibility antigen type,red blood cell type,immunoglobulin allotypes,and isoenzymes,the cells derived from the tu-mor cells can carry on their normal differentiated functions in the chimera.Remarkably,a male tumor mosaic mouse, named Terry Tom,was able to serve as the father of nor-mal offspring that had the genotype of the tumor cells[91]. This is a common property of normal embryonal stem cells, but has been reported only once for teratocarcinoma stem cells.In this case,the transplantable tumor may be consid-ered to be the grandparent of these F1mice.Regardless of the ability of embryonical carcinoma cells to become func-tioning gametes,the differentiation potential of totipotential cells–and in fact their ability to produce cancers–is con-trolled by environmental signals provided by the appropriate normal tissues,as predicted by Cohnheim[65]and Ribbert

[67].Cancer can be viewed as:blocked ontogeny[93,94],

a problem of the control of the progenitor cell by its niche [13,73],or a matter of chemical morphogens altering stem cell differentiation[95].

Transplantable teratocarcinomas are different from other tumor types in that they have no demonstrable chromosomal abnormalities[56]–an impressive fact,in that it suggests that malignancy in not necessarily an inherent property of

10S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–28

a cancer cell.The growth and differentiation characteristics of a cancer are also determined by the relationship of the cancer cell to its environment.The control of the growth of some cancers thus depends on speci?c recognition markers between the cancer cells and the surrounding cells.

4.4.Embryonic cells compared to embryonal carcinoma cells

Embryonic stem cells cultured from the inner cell mass of the early embryo[56,96]are similar in many ways to em-bryonal carcinoma(EC)cells.However,they rarely give rise to tumors,they retain a normal karyotype in vitro,and they routinely retain the potential to contribute to the germ line of chimeras,giving rise to gametes that can transmit the embry-onic cell genotype to the next generation[97,98],a property only once reported for EC cells[56,91,99].Although most of the time the malignant potential of the teratocarcinoma cells in the chimeric mice is controlled by the normal en-vironment,some of these chimeric mice do develop tumors [100].This is not found when embryonic stem cells from the inner cell mass are injected into blastocysts[101].Thus there are differences between the embryonal carcinoma cells derived from germinal stem cells and the embryonic stem cells derived from normal embryos and normal germinal stem cells[99].EC cells may acquire karyotypic abnormal-ities when cultured in vitro,but they are unable to transmit speci?c-induced genetic mutations to chimeric mice[102].

4.5.Regulation of tumor cell differentiation

Is the capacity to induce differentiation of malignant cells a general property of cancer cells,or only of teratocarci-noma tumor stem cells?Injection of leukemia cells into the placenta of a10-day-old mouse fetus induced hematopoietic maturation and the appearance of normal white blood cells carrying the leukemia cell marker in the blood of the mature animal[103].Cells of neuroblastoma[104]and melanoma [105]are similarly regulated if placed in embryonal?elds where normal differentiation of these cell types occurs.Thus, it appears that some tumor cell types in addition to terato-carcinomas can be regulated by the appropriate embryonic ?elds appropriate for a given type of tumor.

Normally during development,both cell proliferation and apoptosis are ongoing as the cells of the embryo interact to form the differentiating organs.Could it be that the process that controls the development of normal cells can also be ap-plied to tumor cells?Using giant blastocysts to obtain blas-tocyst?uid[106],it was found that apopotosis of developing cells is caused by the catabolism of polyamines mediated by enzymes such as amine oxidases.The mechanism of apop-tosis is related to the generation of hydrogen peroxide and a developmentally regulated glutathione-dependent protec-tion mechanism[107],which is active not only in the blas-tocyst,but also in the embryonic limb[108].More recently, the hypothesis that embryonic products extracted from the pregnant uterus or from embryos could affect the growth and differentiation of cancers[109,110]has been tested.In vitro, tumor cell lines(glioblastoma,melanoma,renal adenocar-cinoma,breast cancer and lymphoblastic leukemia treated with extracts from zebra?sh embryos obtained during the differentiation stage induced slowing of proliferation,but not when the extract was taken from a mainly replicative stage [111].The application of this interesting approach remains to be worked out.However,the possibility that immature cancer cells can be induced to differentiate has potential for tumor therapy(differentiation therapy;see below).

5.Skin cancer

5.1.Skin cell lineage and malignant phenotype

The cellular type of a skin cancer depends on the stage of differentiation of the epidermal lineage when the cell under-goes malignant change and maturation arrest(Fig.6,for re-cent reviews see[112,113]).The lineage of the skin includes the precursor cells in the bulge of the hair follicle,more determined basal cells in the epidermis,suprabasal prolif-erating cells(transit-amplifying cells)and non-proliferating cells above the transit amplifying cells.Although this is an oversimpli?cation,transformation of the primitive skin progenitor cells in the bulge of the hair follicle gives rise to trichoepitheliomas,which vary in cellular differentiation but usually contain both keratotic and basal regions,as well as clear cells characteristic of hair follicle[114].Ac-tivation of the more determined basal cells of the skin by overexpression of Ras produces squamous cell carcinoma [115,116],and expression of the c-myc gene in normally non-proliferating suprabasal cells reactivates the cell cycle and leads to hyperplasia(papillomas);these,however,do not progress to invasive tumors[117].Different factors appear to be responsible for growth,e.g.Ras,p53,whereas other factors,e.g.?-cantenin for differentiation[113].Mutation in one gene may not be suf?cient for the malignant pheno-type,but increased proliferation associated with a mutation in a growth regulating gene,associated with a muatation in a differentiation controlling gene,may be suf?cient[112]. In squamous cell carcinoma of the uterine cervix associated with human papilloma virus infection,the initially infected and transformed cell is the basal stem cell,but productive infection requires keratinocyte differentiation[118,119]. 5.2.Skin cancers arise in self-renewing stem cells (initiation and promotion)

Perhaps the best evidence that tumors arise from stem or progenitor cells is provided by the two-step model of skin carcinogenesis[120].The two steps are initiation and promotion[121–124].In the classic model,benz(o)pyrene, the initiator,is painted onto the skin.This chemical binds to DNA in the skin cells,causing a permanent genetic al-

S.Sell /Critical Reviews in Oncology/Hematology 51(2004)1–28

11

HAIR FOLLICULE BULGE CELLS

TERMINALLY

DIFFERENTIATED KERATINIZED CELL

PAPILLOMAS

Fig.6.Skin cell lineage and cancer type.The phenotype of epidermal carcinomas is related to the stage of differentiation of the cell types in the skin where the malignant phenotype is expressed.The most primitive cell is in the bulb of the hair follicle,and the most differentiated cell is the terminally differentiated keratinized cell.

teration (initiation).However,cancers will not arise unless a proliferative stimulus is also given (promotion).This is provided by treating the skin with phorbol ester.Thus,the initiation event induces genetic damage,and the promoter then stimulates the damaged cells to proliferate,leading to cancer.Initiation must occur before promotion.If pro-motion is performed prior to initiation,cancers will not develop.

In the original experiments of Peyton Rous [120,121],initiation was accomplished by painting the ear of a rabbit with coal tar.This produces genetic lesions in the epidermal cells by inducing DNA/carcinogen adducts that cannot be repaired.If no further insult occurs,tumors do not develop.However,if the site is wounded by scraping with a cork borer,epithelial cancer appears at the edge of the wound.The time between initiation and promotion is the critical factor in implicating the stem cell as the initiated cell.This interval can be days,or even months or years in length [122–125].In order for tumors to grow in this model,the initiated cells must survive from the time of initiation to the time of pro-motion.Given the well-established fact that all cells in the skin,except for the self-renewing progenitor cells,turn over completely every 2–3weeks in mice and about 2months in humans [126,127],it is clear that the only way in which the initiated cells could still be present,if months or years have passed since initiation,would be for initiation to have oc-curred in the self-renewing progenitor cell population.In the course of the year or more between initiation and promotion,all of the transit-amplifying cells would have been replaced by newly generated cells from the basal stem cells.Thus,in the initiation-promotion model for skin carcinogenesis,the initiated cell must be a self-renewing progenitor cell.

If this is true,then why don’t the initiated cells grow out and produce cancer during normal tissue renewal?Why is promotion required for initiated cells to grow into cancers?

The answers to these questions are not known.Presumably,normal tissue renewal does not involve proliferation of the long-term self-renewing cells,and any malignant mutations in the transit-amplifying cells will not be expressed as can-cer,since these cells will terminally differentiate.The ini-tiated cell that gives rise to cancer must be a resting stem cell that is only called upon to proliferate under the stress of promotion.

5.3.Field cancerization

The human counterpart of the experimental animal model of initiation and promotion,and the stem cell origin of ep-ithelial cancer,are supported by the phenomenon of ?eld cancerization (Fig.7)[128].Field cancerization was ?rst recognized in humans by the presence of biologically ab-normal tissue surrounding oral squamous cell carcinoma,and the development of multiple primary tumors and locally recurrent cancer in these areas.Field cancerization is asso-ciated with the acquisition of a genetic change in a stem cell,with formation of an altered patch of epithelium which can be recognized histologically by changes such as hyper-keratosis,focal acantholytic dyskeratosis,and acantholysis;these changes re?ect abnormalities of proliferation and/or differentiation of the epithelial cells [129].Patches of ?eld cancerization may have a zone of cells surrounding various epithelial cancers containing mutations of TP53,or showing loss of heterozygosity [130].Thus,?eld cancerization may be considered a manifestation of an initiation event.If fol-lowed by expansion of the initiated population (promotion),a population of cells at high risk for developing further ge-netic changes,leading to invasive cancer is produced.When such a situation is recognized clinically,it is advisable not only to remove any obvious tumors,but also to remove the surrounding “cancerized”?eld,if possible [130].

12S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–28

CANCER e.g. c-myc

Fig.7.Field Cancerization.Epidermal carcinomas are frequently found to be surrounded by a“?eld”of morphologically altered cells.These cells are believed be changed by mutation or loss of a gene such as p53,which produces abnormalities in proliferation.It is postulated that a second mutation, such as in c-myc,then leads to malignant transformation.

6.Liver cancer

6.1.Participation of various cells in the hepatocyte lineage in hepatocarcinogenesis

Cells at various stages of the liver cell lineage proliferate in response to liver cell injury,or in response to hepato-carcinogens[131,132].The?nding that surgical removal of up to two-thirds of the liver was compensated in a few days by proliferation of hepatocytes,with no evidence of involvement of progenitor cells,demonstrates that mature adult liver cells can proliferate[133–135].On the other hand,carcinogenic studies during the1930s and1940s demonstrated that both formation of nodules of hepatocytes (preneoplastic nodules)and hyperplasia of small round cells in the portal zone of the liver preceded hepatocellu-lar carcinoma(HCC)induced by butter yellow[136–139]. The small cells became known as oval cells[140],but most attention was focused on the preneoplastic nodules as the putative precursor to cancer[140–142],suggest-ing that HCC developed from hepatocytes and not from ductal precursor cells.This concept was supported by the conclusion that bile duct cells and hepatocytes were sepa-rate cell lineages[143].The kinetics of the early elevation of the fetal protein,?-fetoprotein(AFP)during chemical hepatocarcinogenesis gave the?rst clue that various cell populations are involved[131,144,145].When the kinetics of serum AFP elevations was followed during the cyclic N-2-acetylamino?uorene(AAF)feeding model of Teebor and Becker[142],AFP became elevated before nodules appeared[146].Localization of the AFP-positive cells and proliferating cells indicated that periductular cells were the ?rst cells to proliferate and to express AFP during carcino-genesis in studies using a choline de?cient diet[147–150]. However,different carcinogenic regimens induced pro-liferation of non-ductular and ductular oval cells preced-ing HCCs[151,152].Then,using a modi?cation of the Solt-Farber[153]model,a number of investigators demon-strated proliferation of oval cells predominately in ducts [151,154–158].A review of the various cellular changes preceding HCC in experimental hepatocarcinogenesis in rats has led to the conclusion that different cells in the liver lineage,including mature hepatocytes,biliary progenitor cells,and periductular cells,can give rise to HCC(Fig.8). The presence of periductular primitive liver stem cells [159],and their early proliferation during hepatocarcinogen-esis[148,149],has stimulated an interest in pleopotenital bone marrow cells as to the possible origin of these cells [132].

HEMATOPOIETIC

STEM CELL

?

Fig.8.Postulated stages of the hepatocytic lineage that may respond to liver injury or carcinogenic protocols.Following various models of liver injury or chemical hepatocarcinogenesis different cell types in the hep-atocytic lineage may respond:1Undifferentiated periductular oval cells, which may arise from circulating bone marrow precursor cells.2.Periduc-tular cells intrinsic to the liver,3.Bipolar ductal progenitor cells,or 4.Mature hepatocytes,which retain the potential to divide.Periductular cells respond to periportal injury induced by allyl alcohol or to choline de?ciency-ethionine carcinogenesis.Bipolar ductal progenitor cells re-spond to injury and to carcinogenic regimens,such as the Solt-Farber model,when proliferation of hepatocytes is inhibited.Hepatocytes re-spond to partial hepatectomy and to carcinogensis by diethylnitrosamine (DEN)(from[132,315]).

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–2813

Given the?nding that hepatocytes can arise from bone marrow precursors[160–163],it seems likely that circu-lating bone marrow-derived stem cells that enter the liver will do so in the periductular region,via the hepatic artery or the portal vein.Is it possible that these circulating bone marrow-derived cells may not only give rise to cells in the hepatocytic lineage,but also to HCC[132]?

6.2.Fusion of bone marrow and liver cells

The possibility that bone marrow stem cells might fuse with recipient liver cells in vivo was raised when it was re-ported that sex mismatched syngeneic bone marrow cells fused with recipient liver cells during restoration of chroni-cally damaged liver in fumarylactoacetate hydrolase(FAH) knockout mice(Tyrosinemia)[164,165].In these recipient mice,there is remarkable ongoing chronic liver damage and proliferation with polyploidy of recipient cells,raising some doubts about the origin of the“fused cells”.There are con-?licting reports on the signi?cance of fusion in normal re-placement of liver cells.Both Newsone et al.[166]and Ishikawa et al.[167]found that human cord blood mononu-clear cells could give rise to human hepatocytes in recipi-ent immune de?cient NOD/SCID mice,with no evidence of fusion.For example,Newsone et al.[166]examined5620 HepPar1(human maker)positive cells in the NOD/SCID mouse liver;each of these cells had nuclear staining char-acteristic of human nuclei and contained only human DNA by FISH analysis.In more sophisticated experiments us-ing cre-lox donor recipient models,both positive and nega-tive fusion results have been obtained.Alarez-Dolando et al. [168]counted both non-fused donor cells and fused cells, but Ianus et al.[169]?nd no evidence of fusion.If fusion does occur,and hepatocellular cancers arise from bone mar-row stem cells that have fused with hepatocytes,this could explain the marked variations in ploidy seen in HCC.

7.Adenocarcinomas

7.1.Stem cell origin

The observations on the stem cell origin of teratocarci-nomas and HCCs may be extrapolated to other organs[6]. Exocrine glands consist of acini of glandular cells drained by a collecting duct.The key progenitor cells are located between the gland and the duct.For example,it is known, both from histologic examination of human lesions[170]and from examination of“preneoplastic”lesions of experimental mouse models[171,172],that breast cancers arise from an undifferentiated suprabasal progenitor cells above the my-oepithelial cells in the duct or the terminal ductular lobular unit.The cells in the terminal lobular unit are the same cells that proliferate during pregnancy and produce progeny that eventually differentiate to form the lactating mammary gland [172].Prostate cancer also arises from suprabasal stem cells [173–175].Intestinal carcinogenesis is directed toward func-tionally anchored stem cells in the crypts[176],and mutated stem cells appear to be able to spread through the mucosa, by fusion of crypts,before becoming invasive[177].Muta-tion in the adenomatous polyposis coli gene(APC)is asso-ciated with increased expression of survivin and?-catenin, which allows expansion of crypt stem cells and development of colon cancer[178].Finally,mixed tumors of epithelial and mesenchymal components(carcinosarcomas)appear to arise from a common monoclonal origin[179].In each in-stance,carcinomas arise from tissue progenitor cells.Is it possible that these cells arise from circulating stem cells that have the ability to cross basement membranes,and to local-ize in various glandular tissues?One of the best examples of the role of maturation arrest of tissue progenitor cells (transit-amplifying cells),due to malignant mutations in the primitive stem cell is that of the hematopoietic system. 8.Hematopoietic stem cells and leukemia

8.1.Normal blood renewal

Malignant cancers of the blood provide one of the clearest examples of the role of stem cells in cancer.Blood is one of the most rapidly replaced tissues in the body[180].In1917, Pappenhein[181]postulated the existence of an undifferen-tiated stem cell for blood cells,“gemeinsame Stammzelle”. These hematopoietic or blood-forming stem cells are located in the bone marrow.The ability of the bone marrow progen-itor cells to form colonies of various types of blood cells in the spleen after transplantation was?rst recognized by Till and McCullough[182]in1961.The frequency of these pro-genitor cells could be estimated from the number of colony forming units[183].By the use of such techniques,it was possible to show that a very small number of progenitor cells can both reproduce and give rise to the complete spectrum of phenotypic colonies of blood cells[184].The number of proliferating hematopoietic progenitor cells has been esti-mated to be0.05%of the total number of bone marrow cells [185];the number of non-proliferating parental stem cells for the proliferating cells is even less.

8.2.Multi/pluripotent hematopoietic stem cells

The multi/pluripotent most primitive hematopoietic cell in the bone marrow is a resting stem cell,which normally does not proliferate,but which can respond to stress,such as the loss of blood or adjustment to a low oxygen environ-ment(high altitude).The primitive hematopoietic stem cell is the parental cell for the major population of proliferating cells in the bone marrow;the transit-amplifying progeni-tor cells(Fig.9;[186]).The progenitor cells,in turn,give rise to progeny that differentiate to mature circulating blood cells[19].Circulating blood cells vary from polymorphon-clear cells,which only live for a day or two,to erythrocytes,

14S.Sell /Critical Reviews in Oncology/Hematology 51(2004)1–28

T R A N S I T A M P L I F I C A T I O N

PLEOPOTENT LONG-TERM STEM CELL (very rare) (CD34-, c-Kit+, Sca1+)

Fig.9.Hematopoietic Cell Lineage.Suggested differentiation pathway of bone marrow stem cells in the hematopoietic pathway.The multi/pluripotent stem cell is a reserve stem cell and normally proliferates very rarely.The expansion of the cell lineage is accomplished by transit amplifying cells that expand into nine different cell lineages.The number of multi/pluripotent stem cells may be very low,whereas the number of various differentiated cell types is very high.

which survive for months,to lymphocytes,which survive for days to years [187].The majority of circulating blood cells cannot proliferate.For example,erythrocytes have excluded their nuclei,and polymophonulclear cells have inactivated their nuclei and condensed them into clumps.Because of the large number of polymorphonuclear cells and their rapid turnover,a very large number of precursor cells is required to generate polymorphonuclear cells.These are present in the bone marrow as transit-amplifying blast cells existing at various stages of differentiation into mature polymorphonu-clear cells.A close relative of the polymorphonuclear cells,the circulating monocytes,are derived from a common pro-genitor cell,but retain the ability to proliferate.8.3.Bone marrow transplantation and pleotypic potential

After bone marrow transplantation,the donor bone mar-row cells can give rise to circulating cells that have the potential to differentiate into unexpected cell lineages (pleopotential),including:endothelium [188–191],muscle [192],liver [157,160–168,193],pancreatic islet beta cells [169,194,195],heart [196,197],brain [198–202],type I [203]and type II pneumocytes [162],kidney parenchyma [204],skin [63,205],ocular retina [206],glomerular en-dothelial cells [207,208],glomerular mesangial cells [209],and other organs [162].Although the bone-marrow cells from which these various cell types have been derived have markers of the hematpoietic stem cell [162,163],it has not been ruled out that this pleopotent cell is of stromal origin [210–212].Serial transplantation indicates that a sin-gle bone marrow cell may give rise to many different tissue

types [57,162]and suggests that a common precursor must exist,not only for the stromal and hematopoietic lineages,but also for other germ layer-derived cell types [187].It is this putative pleopotent bone marrow cell that has stimu-lated the great revival of interest in adult stem cells in the last few years.Although it is dif?cult to prove that there are not different stem cells for different tissues in the bone marrow,at least three studies demonstrates that single bone marrow stem cells can give rise to different epithelial tissue types after serial transplantation [57,162,191].Regardless if there is a single pleopotent cells or multiple stem cells with different potential,the putative stem cells arising from the bone marrow of adults may have just as much promise as embryonic stem cells for stem cell therapy [213–215].8.4.Potentiality caveats

There are some caveats to the generally accepted assump-tion of pleo/multi/pluripotentiality of tissue stem cells,in-cluding hematopoietic stem cells.For example,although it was reported that transplanted bone-marrow stem cells pro-vided new endothelium in transplant models of atheroscle-rosis,more careful analysis in one model suggests that the new endothelial cells were actually derived from pre-existing endothelium [216].In a study of muscle-derived cells con-tributing to bone marrow,the muscle-derived bone marrow stem cells were found to be derived from hematopoietic stem cells present in the muscle [217].Whereas Bjornson et al.[218]generated substantial numbers of hematopoietic pre-cursor cells from neural stem cells,Morshead et al.[219]could not reproduce this result (see Nature Med.8(3002)535–537).In addition,recent studies have shown that in

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–2815

vitro culture of adult mouse brain or bone marrow stem cells with embryonic cells generates hybrid cells containing chro-mosomes of both adult and embryonic cells[220,221],thus raising the question of whether adult stem cells make other tissues by fusing with existing tissue cells.Both of these studies employed co-culture of adult cells with embryonic cells,which is not the case in bone marrow transplants in adults.However,this?nding has stimulated in vivo stud-ies investigating the possibility that transplanted donor bone marrow cells fuse with recipient cells in various organs.As mentioned above,the results are con?icting.In a chronic model of restoration of damaged liver,fusion of bone mar-row cells and liver cells was described[164,165],whereas fusion was not seen in restored pancreatic islet cells[169], in hepatocyes of immune-de?cient(NOD-SCID)mice re-ceiving human bone marrow cells[166,167],or in the buc-cal mucosa of female recipients of male bone marrow cells [222].The demonstration of fusion in the chronic severe in-jury models may be due to marked disruption of the archi-tecture of the liver and compression of the circulating cells against damage cells.Thus,the possibility of fusion in vivo, in the absence of extensive cell injury or in mature diploid cells[57],remains to be resolved(see[223]).

8.5.Unanswered questions

Assuming that pleopotent bone marrow stem cells actu-ally exist,a number of basic questions remain unanswered. Is there a single cell type in the bone marrow that is pleopo-tent,or are there various types of“progenitor”cells that can give rise to different cell types in other organs?It appears that a single cell that can do it all[57,162,191].Is the abil-ity of a bone marrow stem cell to contribute to an epithe-lial organ a very rare or a more common event?Krause and colleagues[57]?nd it to be a fairly common occurrence; whereas Wagers and colleagues[224]claim that this is an extremely rare event.The ability of bone marrow-derived cells to contribute to epithelial tissue may depend on the age of the donor and the method of puri?cation of the stem cells,but clearly the weight of published evidence in favor of a pleopotent bone marrow stem cell outweighs the nega-tive evidence[225].Does the progenitor cell normally cir-culate,or is it liberated from the bone marrow by humoral signals after injury?The evidence is that this cell circulates normally,but that its numbers may be increased after injury. Do progenitor cells normally exist in mature organs,such as liver or kidney,and can they re-circulate back to the bone marrow?Are these cells derived from the bone marrow,or can totipotent progenitor cells be generated in differentiated organs[226]?The ability of brain cells to become blood cells[218,227]suggests that the answer to the latter ques-tion is“yes”,but re-circulation of blood-derived cells to the brain,and back to the bone marrow,cannot be ruled out. Can progenitor cell lines that could be used for transplanta-tion,for organ replacement,or for delivery of gene therapy, be derived from bone marrow or other organs[187]?Ex-tensive culture of neural stem cells may be required before such cells can differentiate into blood cells,suggesting that rare differentiation events may occur upon culturing;and that these events may account for neural to blood differen-tiation[219].This?nding suggests that normal brain pre-cursor cells cannot contribute to blood-cell lineages in vivo. Answers to these questions,and clinical applications of the use of tissue stem cells,will undoubtedly be forthcoming in the next few years[187,225].

9.Leukemias as models of maturation arrest of a cell lineage

9.1.Leukemia as a stem cell cancer

Leukemias are caused by a genetic change or in a single cell that produces progeny that continue to proliferate,and/or do not die[228].The demonstration that tumor growth de-pends on a subpopulation of proliferating stem cells(cancer stem cells)in the tumor was?rst described in transmissi-ble leukemias of mice.Furth and Kahn[229]in1937were able to transplant leukemia from one mouse to another us-ing a single undifferentiated cell.In1955,Makino and Kano [230]obtained clones of tumor cells from single cells.Hu-man leukemias feature gene rearrangements that are present in all the cells of the tumor[231,232],suggesting the tumor’s origin in a single progenitor cell that has undergone a ma-lignant gene rearrangement.This concept also holds true for leukemias and lymphomas of humans(for a recent review, see[233]).The effect of a genetic change in the precursor cell of the population is exempli?ed by the malignant in-crease of multiple cell types in chronic mono/myelogenous leukemias,including various types of polymorphonuclear cells(neutrophils,eosinophils,and basophils),as well as monoctyes,erythrocytes,and platelets(megakaryocytes),all of which contain the same genetic lesion or lesions.Thus, the malignant cell is the multi/pluripotent tumor stem or pro-genitor cell with the capacity to differentiate into mutiple types of blood cells[234].

9.2.Chemotherapy and reserve stem cells

The pattern of response to chemotherapy of acute leukemias also implies that these cancers arise from a prim-itive stem cell[234].Repeated cycles of anti-proliferative treatment are required to affect resting tumor cells that are not cycling at one speci?c time of therapy.Since the ther-apy acts on actively proliferating cells,one cycle of drug treatment should be suf?cient to eliminate the actively pro-liferating cells present at the time of treatment.However, the malignant genetic change is also present in the“resting”or G0stage stem cells that are not proliferating when the drug is given.Successful treatment may require four or more cycles of anti-proliferative therapy to catch all of the leukemic cells.Even so,up to30%of patients will require

16S.Sell /Critical Reviews in Oncology/Hematology 51(2004)1–28

Table 2

Some gene rearrangements in leukemias and lymphomas Leukemia/lymphoma Gene rearrangement Gene activated

CML t9:22bcr/abl Tyrosine phosphorylase AML t8:21IL-3R

Tyrosine kinase

APL t15:17PML/RAR ?Retinoic acid receptor,blocks diff.ALL

t12:22TEL/MN1Transcription factors t9:12TEL/abl Tyrosine phosphorylase Burkitt’s (B-cell)t8:14IgG/myc c-myc (transcription)B-cell lymphoma t14/18IgG/bcl2bcl-2,blocks apotosis CLL

?

?

blocks apotosis

Listed are only a few of the many gene rearrangements found in leukemias and lymphomas.These rearrangements frequently result in translocation of a promoter and a structural gene,resulting in activation of expression of the rearranged gene.The two major classes are those that activate kinases and cause proliferation,and those that activate genes (such as bcl-2),that block apoptosis.Examples of how these gene rearrangements are manifested in leukemias and lymphomas are presented in Figs.10and 11.

total bone marrow ablation and stem cell transplantation from a normal donor,in order to eliminate all of the primi-tive resting cells that have the genetic lesion.This implies that the leukemia arises in the reserve hematopoietic stem cell,which can only be eliminated by total bone marrow ablation.

9.3.Gene translocations and leukemias

The characteristics of various leukemias illustrate the relationship between the stage of maturation arrest of the cells and the genetic change;this determines the natural history (growth rate)of the tumor.A few of the known

gene

EOSINOPHILS NEUTROPHILS BASOPHILS

, IL-6

IL-5LIF

ERYTHROCYTES

Fig.10.Constitutive activation of the IL-3receptor causes myelogenous leukemia.IL-3is active in the proliferation and differentiation of all cells in the myeloid series,including polymophonuclear leukocytes,monocytes,megakarocytes (platelets),and erythrocytes.Thus,constitutive activation of the IL-3receptor results in an increase in the precursors of all of these cell types,a characteristic of acute myelogenous leukemia.Modi?ed from [316].

rearrangements in leukemias and lymphomas are listed in Table 2.Leukemias feature genetic lesions that affect either cell proliferation (transactivation)or cell death (apoptosis).The nature of the genetic lesion can be directly related to the type of leukemia [228,231,235,236].

9.3.1.The IL-3receptor

In acute myelogenous leukemia (AML)there is often a gene rearrangement leading to constitutive activation of the IL-3receptor resulting in activation of tyrosine kinase and cell proliferation.The role of IL-3in normal hematopoiesis is shown in Fig.10.From this ?gure it can be appreci-ated that activation of the IL-3receptor should produce

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–2817

Fig.11.Activation of immunoglobulin promoter linked to oncogene(c-Myc)and/or apoptosis blocking gene(Bcl-2)produces B-cell cancer in transgenic mouse model of Burkitt’s lymphoma.B-cell lymphomas occur when the immunoglobulin gene promoter is linked to an oncogene(c-Myc)and/or a gene that codes for the apoptosis blocker,Bcl-2.Although the Ig-promoter/oncogene translocation is present in every cell in the animal,the promoter is only activated in B-cells,so that other cells in the lymphocytic lineage are not affected.

an increase in all cells in the myeloid lineage,the charac-teristic of AML.With respect to determining the stage of differentiation at which the genetic rearrangement is found, the rearrangement is generally identi?able in the early myelogenous progenitor or even in the more primitive bone marrow stem cells(see above).

9.3.2.B-cell lymphomas and the immunoglobulin heavy chain gene promoter

The role of translocation of the immunoglobulin(Ig)gene promoter with either activating or apoptosis-blocking genes for B-cell lymphoma is illustrated in Fig.11.In transgenic mice,translocation of the Ig-gene promoter to the c-Myc and/or Bcl-2gene may produce a B-cell lymphoma[237] similar to Burkitt’s lymphoma[238].In this model it is pre-sumed that the genetic lesion is present in all cells.However, the effect of the gene is not manifested until the Ig-heavy chain gene promoter is activated in B-cells.Thus,activation of the oncogene c-Myc or the apoptosis blocker Bcl-2linked to the Ig promoter does not occur until the Ig-gene promoter to which they are linked is activated.When this occurs,there is either increased proliferation(c-Myc)or decreased apop-tosis(Bcl-2),or both,resulting in a rapid increase in the number of immature B-cells that do not die.Thus,the ge-netic lesion causing leukemias occurs in the stem cells,but the growth characteristics of the tumor are determined by the stage of maturation of the B-cell lineage in which the Ig-heavy chain gene promoter is activated.

9.4.Stage of maturation arrest and clinical course of leukemias

The degree of differentiation and the clinical behavior of a cancer are directly related to the stage of maturation arrest of the cells in the tumor lineage(Fig.4).The clinical course of a given leukemia depends on the rate of cell proliferation (growth fraction)and the rate of cell death.A high percent-age of large blast cells is a characteristic of fast-growing acute leukemias,with little or no terminal differentiation, whereas a high percentage of mature differentiated cells is a characteristic of chronic leukemia(Fig.12).Blast cells represent cells that are in the cell cycle;mature cells repre-sent cells that are not in the cycle.A leukemia composed of blast cells has a high growth fraction(a high percentage of cells in cycle at any given time).A leukemia composed of mature cells,as in chronic lymphocytic leukemia,has a low growth fraction.A mixture of blast and more mature cells implies an intermediate growth fraction.In chronic lympho-cytic leukemia(CLL)most of the cells are resting G o lym-phocytes,with fewer than1in1000cells in cycle at any time,but the cells of CLL do not die,and instead accumu-late over years.

The clinical course of leukemia is determined by the rate of replacement of the normal bone marrow progenitor cells with leukemic progenitor cells.This replacement results in a loss of the normal cells and their function.Loss of red blood cells results in anemia;loss of platelets leads to bleeding; loss of polymorphonuclear cells causes decreased resistance to infection.Leukemia with a high growth fraction will re-place the normal progenitor rapidly(in months),whereas leukemia with a low growth fraction may take years to pro-duce symptoms(Fig.12).Thus,the histologic classi?cation and the clinical course of leukemia are determined by the stage of arrest of differentiation where the genetic lesion is activated.Depending on when the lesion was acquired,it may be presumed that the genetic lesion is present in many other cells of the body,including the pleopotent bone mar-row stem cell,but the malignant phenotype is only expressed when the genetic lesion is activated.If it is activated early in the course of maturation,most of the cells will re?ect the properties of actively dividing transit-amplifying cells, such as in the multi/pluripotent hematopoietic progenitor

18S.Sell /Critical Reviews in Oncology/Hematology 51(2004)

1–28

Fig.12.Growth fractions,morphology,and clinical course of three selected leukemias.In acute myelogenous leukemia,the tumor cells are arrested in an active growth phase:cells that divide and do not enter G 0,but pass directly into the next cell cycle.Few,if any,differentiated cells are seen;the tumor is made up of blast cells.The growth fraction is very high,the expansion of cells is essentially exponential,and the time to death,if the disease is not treated,is within a few months.In chronic myelogenous leukemia,the arrest is at the level of the transit-amplifying cells.The number of cycling cells is much smaller,and a much higher proportion of the tumor cells undergoes differentiation so that many cells at various stages of differentiation are seem.The growth fraction is small,and the time to death is years.In chronic lymphocytic leukemia,maturation arrest occurs in small non-dividing cells.The functional change is a lack of cell death.The vast majority of the cells are in G 0,so that the growth fraction is vanishing small,and the time to death is decades.Modi?ed from [317].

cells of acute myelogenous leukemia.If the genetic lesion is manifested at a more mature stage of maturation,a chronic leukemia such as CLL will result.

10.Differentiation therapy

If the malignant cells of cancers are cancer stem cells [6,180,233,239–241],then it should be possible to treat can-cers by inducing differentiation of the stem cells,i.e.,dif-ferentiation therapy [242].As discussed above,the tumor stem cells of teratocarcinomas can be in?uenced by the en-vironment of the developing embryo to differentiate into normal adult tissues.If tumor cells can be forced to dif-ferentiate and to cease proliferation,then their malignant potential will be controlled.Although a number of agents have been studied over the years [243],the most thoroughly examined and clinically tested as a differentiating agent is retinoic acid (RA,Vitamin A),in particular all-trans-retinoic acid (ATRA).

10.1.Retinoic acids and teratocarcinomas

Ironically,although teratocarcinomas were the ?rst tu-mors that were shown to differentiate in vitro after addition of low concentrations of RA [244],this has not proven to be an effective treatment in vivo.Addition of retinoic acid (RA)or RA in combination with dibutyryl-cAMP,stimulates dif-ferentiation of a cell line of nondifferentiatied,immortalized embryonal carcinoma cells,called F9,to an epithelial cell type consistent with extra-embryonic endoderm [244–246].RA induces differentiation through activation of speci?c nuclear receptors,RAR,which activate c-Fos/c-Jun medi-ated transcription [247,248],resulting in upregulation of the expression of differentiation genes [249–251](Fig.13),including AFP [252].At least three nuclear retinoic acid

S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–2819

Signaling/

Induces Senescence

Inhibits Proliferation

Fig.13.Retinoic acid(RA)reacts with nuclear receptors in embryonal carcinoma cells leading to inhibition of proliferation and induction of proliferation.RA and related compounds pass into the nucleus where they react with nuclear receptors(RAR-RXR)that from a transcription complex with c-Fos,c-Jun,AP-1,and most likely other activation proteins resulting it upregularion of a number of genes related to inhibition of proliferation and induction of differentiation.

receptors(RAR)-?,-?,and-?,and three retinoid X re-ceptors(RXR)-?,-?,-?have been identi?ed.The most active receptor appears to be a heterodimer between RAR and RXR[253].The RAR-RXR heterodimer binds to DNA sequences called retinoic acid response elements found in the promoter region of target genes and enhance gene transcription after binding their ligands[254,255].A num-ber of cellular pathways are affected during RA-induced activation of RAR.A speci?c candidate tumor suppressor gene product,Disabled-2,is induced after RA treatment. Disabled-2greatly decreases c-Fos expression and disas-sociates MAPK activation,resulting in differentiation of the cells[256].RA-induced differentiation correlates with accumulation of the cyclin-kinase inhibitors p21/Waf1and p27/Kip[257],a decrease in cyclin D3[258],and activation of the?-catenin/Lef-Tcf signaling pathway[259].In addi-tion,RA-induced differentiation of human teratocarcinoma cells leads to increased methylation of the hTERT promoter and silencing of the hTERT gene,which codes for the catalytic subunit of telomerase[260].Differentiation of F9 cells is also induced by depleting putrescine and spermidine, required for polyamine synthesis[261].Thus,RA activates multiple pathways leading to decreased proliferation and increased differentiation of embryonal carcinoma cells. However,treatment of teratocarcinomas that demonstrate sensitivity of cells in vitro to retinoids,has not yielded sat-isfactory results in vivo[262].Some cells may differentiate, but the malignant core of cells remains and quickly grows back.The treatment of choice for human teratocarcinoma is surgery and,if total resection is not possible,follow up treatment with irradiation and/or cisplatin-based cytotoxic chemotherapy[263].The ef?cacy of the treatment is re-lated to the initial tumor burden.Almost80%of patients with germ cell tumors treated with cisplatin remain relapse free for2years,but successful treatment of recurrent dis-ease also depends on tumor burden[264,265].Combination chemotherapy that includes other drugs,such as taxol,may have an effect on refractory cases[265,266].Treatment of refractory testicular cancer with trans-retinoic acid has not proved to be effective even in combination with cisplatin [267].

10.2.Retinoids and acute promyelocytic leukemia(APL) APL is a cancer of children wherein the precursor cells of the myelocytic lineage,expressing markers of the M3type, fail to mature,accumulate in the bone marrow and blood in enormous numbers,and eventually force out production of normal blood cells.The molecular lesion in APL is a t(15:17)gene rearrangement involving the nuclear retinoid acid receptor alpha(RAR?).In this rearrangement the gene for a protein called promyelocytic leukemia protein(PML) is fused to the RAR?gene(PML-RAR?).PML is normally found in a discrete,circumscribed nuclear structure called a nuclear body in cells of the myeloid series[268,269].The PML-RAR?fusion product inhibits the RAR?[270],dis-rupts the nuclear bodies,and[271],and without its ligand, retinoic acid,the fusion product functions as a constitutive transcriptional repressor,blocking hematopoietic differen-tiation and allowing the accumulation of poorly differen-tiated leukemia cells[272].On the other hand,the APL fusion gene product appears to induce genes that maintain the stem cell phenotype and represses DNA repair genes, resulting in a mutator phenotype that enhances tumor pro-gression[273].ATRA treatment upregulates the ubiquitin activating enzyme-E1like protein(UBE1L)that triggers degradation of the PML/RAR?fusion protein[274],acti-vates RAR?mediated transcription,allows reformation of the PML-nuclear body and stimulates differentiation and eventual apoptosis of APL cells[271,275].

Currently about90%of newly diagnosed patients with APL achieve complete remission and over70%are cured by ATRA therapy[276]with or without concomitant chemotherapy with methotrexate and cytarabine[277].The clinical presentation of APL is frequently associated with hemorrhage and low platelet counts due to the decreased ability of the bone marrow to produce platelets.Poor prog-nostic factors include older age,elevated white blood cell count,low platelets,and CD56expression[278].The overall survival rate is greatly increased by ATRA versus chemotherapy alone and there is an up to75%disease free ?ve year survival rate for both induction and maintenance on ATRA treatment[279].After achievement of complete remission,maintenance therapy with ATRA with low-dose chemotherapy is may be useful.Despite overall great suc-cess in treating APL with retinoids,relapse with develop-ment of acquired resistance to retinoid induced maturation is not uncommon and is responsible for treatment failure [280,281].The mechanism of resistance to RA therapy is not well understood.It may be due to increased ATRA metabolism,increased expression of the RA binding pro-teins,P-gylcoprotein expression,or mutations in the ligand binding domain of RAR-?[282].Due to severe side ef-fects of ATRA treatment(see below)dosing schedules are

20S.Sell/Critical Reviews in Oncology/Hematology51(2004)1–28

critical to the success of treatment.Treatment with other differentiating-inducing agents,cytotoxic or chromatin re-modeling agents,as well as receptor-selective and modi?ed retinoids may overcome this resistance[280].Cytotoxic treatment with arsenic trioxide is the treatment of choice for refractory ATRA resistance[277,278].In addition,re-sistance can be overcome to some extent by combining histone deacetylase inhibitors,such as sodium butyrate, with RA.This treatment facilitates RA-induced gene tran-scription and induction of apoptosis,but has little effect on granulocytic differentiation of the promyelocytic leukemia cells[283].Bone marrow transplantation may be important in the future for patients in relapsed/refractory disease or those in a second CR[278].

10.3.Retinoid differentiation therapy;other cancers

In addition to embryonic cells,teratocarcinomas,and leukemias,RA induces differentiation in other cell types [278,284–288].In vitro,retinoids reduce the ability of human melanoma cells to form colonies[289]and this inhibition correlated in a small number of patients with a modest clinical response[290].Transfection of RAR into non-responsive cells in vitro can restore responsiveness to retinoids by antagonizing activator protein-1(AP-1), blocking proliferation and activating differentiation[291]. Retinoids may be able to upregulate expression of p53 and induce p53dependent G1arrest and apoptosis of human lung cancer cells[292].In solid cancers RA in-creases differentiation and apoptosis,and decreases pro-liferation(blockade of G1),invasiveness,and metastasis [274],Retinoids have been employed with limited success in treatment of many different solid cancers[287],includ-ing squamous cell carcinoma[293],adenocarcinoma of the prostate[294,295],neuroblastomas[296],HCC[297]in humans,and breast carcinoma in mice[298].Treatment of squamous tumor cell lines with ATRA may act though the RAR to upregulate a putative class II tumor suppres-sor gene,tazarotene-induced gene3(TIG3)[299].13-cis RA may be used in conjunction with other agents,such as inferferon-?,?-tocopherol and cisplatin,for treatment of advanced squamous cell carcinoma[300,301].After ini-tial clinical remission,solid tumors,like APLs,frequently develop resistance to retinoid-induced maturation[282]. 10.4.Chemoprevention of cancer by retinoids

If retinoids can induce differentiation of malignant tumor stem cells,then it is even more likely that they could in-duce differentiation in so-called“premalignant”cells and be used as chemopreventive agents for cancer[287].In addition to the premalignant changes seen in the bronchial epithe-lium of smokers,RA has been used with some success to cause regression of laryngeal papillomatosis and oral leuko-plakia[302].Loss of nuclear retinoid acid receptor(RAR)in bronchial epithelium is considered a marker for preneoplasia frequently found in smokers and an indicator of increased risk for lung cancer[303].Many premalignant and malig-nant cells have a reduced expression of RAR?,and treatment with retinoids may induce RAR?and induce differentiation in some cancer cell lines[292]and in biopsy specimens of bronchial epithelium of smokers[304].Treatment with 9-cis-RA can restore RAR expression in former smokers, raising the possibility that this could be a chemopreventive agent for lung cancer[305].In an earlier study,treatment of squamous cell cancers and preneoplastic lesions with retinoids had only a modest clinical effect,and these studies were limited by dose-related toxic effects,including dermati-tis,emotional liability and headache[306].More sophisti-cated studies using different retinoids and other inhibitors, such as growth factors receptor,are proposed([307],but a statistically signi?cant chemopreventive effect has not yet been demonstrated.

10.5.Retinoic acid syndrome

Finally,it seems that there is a price to pay for any approach to cancer therapy.Many patients treated with all-trans retinoic acid develop“retinoic acid syndrome”[308].This syndrome consists of fever,weight gain,respi-ratory distress,interstitial pulmonary in?ltrates,pleural and pericardial effusion,episodic hypotension,and acute renal failure.Recently the recommended dosage of ATRA used for treatment of APL has been reduced from45to25mg/m2 day in an attempt to limit these side effects[309].When identi?ed early,this syndrome can be effectively treated with dexamethasone[288].

11.Summary and conclusions

It is the hypothesis of this paper that cancers arise from stem cells in adult tissues.All adult tissues are made up of lineages of cells consisting of tissue-determined stem cells and their progeny(transit-amplifying cells),that provide cellular lineages producing many cells that differentiate to the mature functioning cells of the tissue.Each tissue has an individual rate of normal tissue renewal.Tissues with a rapid turnover of cells,such as blood and gastrointestinal lining epithelium,have a relatively large number of imma-ture transit amplifying cells.Other tissues,such as the liver or brain,with a very low normal renewal rate,have very few transit-amplifying cells.In any case,all tissues have stem cells,but these stem cells do not all have identical pos-sibilities(potentials)for producing different cell types.For example,the bone marrow may contain a stem cell that can unexpectedly give rise to various tissue types including ep-ithelial and neural tissues(pleopotent),as well as a stem cell that give rise to blood cells(multi/pluripotentent).The gas-trointestinal lining contains stem cells that can give rise to at least four different cell types(oligopotent),whereas the liver stem cells appear to give rise to two cell types(bipotent).

干细胞存储行业研究报告精编版

2017年干细胞存储行业研究报告 因为具有潜在的再生能力,干细胞自其诞生之日起,就被认为可能是革命性的医疗技术。2007年和2012年的诺贝尔生理或医学奖,分别授予了与干细胞有关的发现。 2011年开始,干细胞概念再次被市场热炒。根据Market Research及Transparency Market Research预计,到2018年全球干细胞储存市场容量将增长至181.6亿美元。 干细胞存储业务的想象空间虽然不及干细胞治疗,但依然是现阶段许多干细胞公司的重要业务。对投资机构来说,一方面存储业务充沛的现金流可以缓解研发费用的压力,一方面细胞分离等技术能力的高低也能侧面反映公司的技术水平,对于分析标的的风险和潜力意义重大,值得持续关注和研究。 一、概述 干细胞是具有自我更新、高度增殖、多项分化潜能及良好组织相容性等特点的细胞群体。因为具有潜在的再生能力,干细胞自其诞生之日起,就被认为可能是革命性的医疗技术。2007年和2012年的诺贝尔生理或医学奖,分别授予了“在利用胚胎干细胞引入特异性基因修饰的原理上的发现”和“发现成熟细胞可被重写成多功能细胞,细胞核重编程技术(iPS细胞)”两项与干细胞有关的发现。 2011年开始,干细胞概念再次被市场热炒。在A股市场上,以中源协和为代表的企业当年股价飞涨。 存储业务涉及的主要是造血干细胞和间充质干细胞,前者主要来源于骨髓、外周血、脐带血、胎盘等,后者主要来源于骨髓、脐带、脂肪、脐带血、羊膜、胎盘等,进而形成了脐带血造血干细胞等细分种类。目前干细胞存储的主要市场仍在新生儿相关的业务,随着分离等技术的成熟和临床研究的深入,新生儿干细胞存储种类正在从脐带血造血干细胞转向胎盘、脐带等来源的间充质干细胞。

win7系统动态桌面设置全攻略

Win7系统动态桌面设置全攻略 您是否厌倦了不会动的电脑桌面?即使是Win7系统自带的自动切换壁纸功能,时间长了也觉得没什么新意,无非就是像幻灯片一样切换而已。实际上,Windows 7 还有一个更棒的隐藏功能——可以将任何WMV格式的视频设置为电脑桌面并循环播放,而且没有时间长度的限制! 试想,当你的同事、家人或朋友路过你的电脑时,发现你的桌面竟然会动!而且呈现的是唯美细腻的高清视频!他们的脸上会是什么表情?从今以后,他们对你的敬仰,一定会犹如滔滔江水,连绵不绝...... 打开这一隐藏功能的方法其实很简单,只需对着屏幕喊:“芝麻开门!桌面动起来!”您的图片壁纸就变成更为生动的视频壁纸了。。。呵呵,开个玩笑,后面我们会告诉您正确方法。当然,Windows 7 系统也确实有语音识别功能,而且挺有意思的,有兴趣的朋友可以点击阅读《Win7真好玩:超有趣的"语音识别"功能》一文。 点击图片查看下一页

动态GIF图,图片打开速度较慢,请稍等 在开始介绍设置方法之前,先请您看看动态桌面的最终效果,然后您再决定是否继续看下去: 视频:动态桌面的实际效果 怎么样,感觉如何?如果您也觉得这个功能很炫,愿意尝试一下的话,请看下面的图文教程,其实非常简单,正常情况下只需两分钟就可以搞定了。

动态桌面补丁的下载和设置过程 言归正传,打开这一隐藏功能只需一个小补丁,它叫做“Windows7-DreamScene”,我们去百度上搜这个名字或是“动态桌面”都能找到,它的体积很小,只有221KB而已。 如果您是保存到桌面上的话,下载完毕后桌面上就会多出一个名为 Windows7-DreamScene.RAR的压缩文件,我们将其解压后,就可以看到这个小补丁的本体了。双击它,就会打开下面这个窗口: 看到这个窗口后,随便按键盘上的一个键即可,比如回车键按任意键后,这个小窗口的内容会不断发生变化,我们不用管它,大概20秒后变化完全停止,我们再按任意键,这个窗口就会消失,暗示我们该补丁已安装成功,Win7的隐藏功能已被打开。

干细胞与组织工程

干细胞与组织工程 随着生命科学的飞速发展,目前组织工程、干细胞研究已经成为21世纪生命科学研究的焦点和前沿领域。组织工程研究涉及种子细胞、生物支架材料以及组织构建等众多研究方向.干细胞研究则有望解决组织工程研究中的种子细胞来源问题,可能成为组织工程研究中的理想种子细胞。 一“组织工程”的概念 1 “组织工程”的产生和发展 组织、器官的损伤或功能障碍是人类健康所面临的主要危害之一,也是人类疾病和死亡的最主要原因。据美国的一份资料显示,每年有数以百万计的美国人患有各种组织、器官的损伤或功能障碍,每年需进行800万次手术进行修复,年住院日在4000万~9000万之间,年耗资超过400亿美元。 随看现代外科学的发展,人类对组织、器官缺损的治疗有了很大的进步,但仍然存在许多问题。目前临床常用的治疗方法有三种: 1.自体组织移植、 2.异体组织移植、 3.人工合成组织代用品 组织工程是近年来正在兴起的一门新兴学科,1984年, Wolter首先提出“组织工程”(Tissue Engineering)一词。1987年,美国国家科学基金会于正式提出和确定“组织工程”一词,开辟了组织工程学研究的新纪元。它是应用生命科学和工程学的原理与技术,在正确认识哺乳动物的正常及病理两种状态下结构与功能关系的基础上,研究、开发用于修复、维护、促进人体各种组织或器官损伤后的功能和形态生物替代物的科学。 从事组织工程研究的科学家们利用细胞生物学、分予生物学以及材料科学等学科的最新技术,像工厂生产零部件一样,针对患音组织或器官缺失情况,利用构成组织或器官的基本单位——细胞以及为细胞生存提供空间的支架材料,在体内外培育出所需的人体组织或器官.需要多少就培育多少.量体裁衣制备完成后再给患者安装上去。 组织工程研究的核心是建立由细胞和生物材料构成的三维空间复合体。这一三维的空间结构为细胞提供了获取营养、气体交换、排泄废物和生长代谢的场所,

柳宗元驳复仇议翻译优选稿

柳宗元驳复仇议翻译集团公司文件内部编码:(TTT-UUTT-MMYB-URTTY-ITTLTY-

臣伏见天后时①,有同州下邽人徐元庆者②,父爽为县尉赵师韫所杀③,卒能手刃父仇,束身归罪④。当时谏臣陈子昂建议诛之而旌其闾⑤,且请“编之于令,永为国典。”臣窃独过之⑥。 注释: ①伏见:旧时臣下对君主有所陈述时的表敬之辞,可译为知道,了解。天后:即武则天(624-705),名武瞾。690年,废睿宗李旦自立为皇帝,改洛阳为神都,建立武周王朝,在位十六年。705年,武则天病笃,宰相张柬之发动兵变,迫使武氏退位,史称神龙革命。中宗李哲复位,恢复唐朝。 ②同州:唐代州名,今陕西渭南市大荔县一带地区。下邽(guī):县名,今陕西省渭南县。徐元庆:当时某驿馆的服务人员,徐元庆替父报仇,谋杀官员赵师蕴案是武则天时轰动一时的谋杀案。 ③县尉:县令的属官,专司当地的治安工作。或称御史大夫。 ④卒:最后,最终。束身归罪:自首。 ⑤陈子昂:(661—702),字伯玉。武后时曾任右拾遗,为谏诤之官。旌(jīng):表彰。闾:里巷的大门。 ⑥过:错误,失当。 译文:微臣知道则天皇后时,同州下邽县有个叫徐元庆的人,他的父亲徐爽被县尉赵师韫杀害,他最后能亲手杀掉他父亲的仇人,并且自己捆绑着身体到官府自首。当时的谏官陈子昂建议将他处以死罪,同时在他的家乡表彰他的行为,并请朝廷将这种处理方式“编入法令,永远作为国家的法律制度”。臣私下认为,这样做是不对的。

臣闻礼之大本,以防乱也。若曰无为贼虐①,凡为子者杀无赦。刑之大本,亦以防乱也,若曰无为贼虐,凡为治者杀无赦。其本则合,其用则异。旌与诛莫得而并焉。诛其可旌,兹谓滥,黩②刑甚矣。旌其可诛,兹谓僣③,坏礼甚矣。果以是示于天下,传于后代,趋义者不知所向,违害者不知所立,以是为典可乎?盖圣人之制,穷理以定赏罚,本情以正褒贬,统于一而已矣。 注释: ①贼虐:残害,践踏 ②黩(dú)刑:滥用刑法。黩,轻率。 ③僭(jiàn):越过,超出本分。 译文: 臣听说,礼的根本作用是为了防止人们作乱。意思是说,不要让礼受到践踏,凡是作儿子的,为报父仇而杀了人,就必须处死,不能予以赦免。刑法的根本作用也是为了防止人们作乱。意思是说,不能让刑受到践踏,凡是当官的错杀了人,也必须处死,不能予以赦免。礼和刑的根本目的是一致的,但是实际应用却不同。表彰和处死是不能同施一人的。处死可以表彰的人,这就叫乱杀,就是滥用刑法太过分了。表彰应当处死的人,这就是过失,破坏礼制太严重了。如果以这种处理方式昭示天下,并传给后代,那么,追求正义的人就不知道前进的方向,躲避刑罚的人就不能辨别立身之道,以此作为法则行吗?圣人制定礼法,是透彻地探究事理来制定赏罚,根据事实来确定奖惩,不过是把礼和刑二者结合在一起罢了。 向使刺谳①其诚伪,考正其曲直,原始而求其端②,则刑礼之用,判然离矣。何者?若元庆之父,不陷于公罪,师韫之诛,独以其私怨,奋其吏气,虐于非辜;州牧不知罪,刑官不知问,上下蒙冒③,吁号不闻。而元庆能以戴天④为大耻,枕戈⑤为得礼,处心积虑,

干细胞行业分析

干细胞行业分析报告 贵州省科技风险投资管理中心 贵州省科技风险投资有限公司 贵州鼎信博成投资管理有限公司

目录 一、干细胞概述 (3) (一)产品定义 (3) (二)干细胞产品分类 (3) 二、干细胞行业及发展概况 (5) (一)国际行业发展概况 (5) (二)国内行业发展概况 (6) (三)干细胞行业特点 (8) 三、国内外干细胞临床研究的概况 (10) (一)国外干细胞临床研究现状 (10) (二)国内干细胞临床研究现状 (10) 四、干细胞市场现状分析 (11) 五、干细胞市场供需分析 (16) 六、涉及干细胞技术研究与应用的上市公司基介绍 (21) 七、干细胞行业产业链及投资价值分析 (22) 附件一:2011年最新国外干细胞研发企业及其相关产品 (25) 附件二:干细胞产业政策研究 (33) 附件三:人类干细胞之伦理原则与监管政策 (35)

一、干细胞概述 (一)产品定义 干细胞(stem cells,SC)是一类具有自我复制能力(self-renewing)的多潜能细胞,在一定条件下,它可以分化成多种功能细胞。干细胞(Stem Cell)是一种未充分分化,尚不成熟的细胞,具有再生各种组织器官和人体的潜在功能,改变了人类疾病的应对方法,医学界称为“万用细胞”。 (二)干细胞产品分类 1、干细胞具有自我更新能力(Self-renewing),能够产生高度分化的功能细胞。干细胞按照生存阶段分为以下五类: (1)胚胎干细胞 胚胎干细胞当受精卵分裂发育成囊胚时,内层细胞团(Inner Cell Mass)的细胞即为胚胎干细胞。胚胎干细胞具有全能性,可以自我更新并具有分化为体内所有组织的能力。 (2)成体干细胞 成年动物的许多组织和器官,比如表皮和造血系统,具有修复和再生的能力。成体干细胞在其中起着关键的作用。在特定条件下,成体干细胞或者产生新的干细胞,或者按一定的程序分化,形成新的功能细胞,从而使组织和器官保持生长和衰退的动态平衡。

免疫学的临床应用

免疫学的临床应用有两个方面:一是应用免疫理论来阐明许多疾病的发病机制和发展规律;二是应用免疫学原理和技术来诊断和防治疾病。本章内容主要是后者。此外,免疫学不仅应用于传统的传染病中,而且在肿瘤、自身免疫病、免疫缺陷病、器官移植、生殖免疫等中均广泛应用。 免疫学防治是指应用免疫制剂或免疫调节药物调整机体的免疫功能,对疾病进行预防和治疗。特异性免疫的获得方式有自然免疫和人工免疫两种。自然免疫主要指机体感染病原体后建立的特异性免疫,也包括胎儿或新生儿经胎盘或乳汁从母体获得抗体而产生的免疫。人工免疫则是人为地使机体获得免疫,是免疫预防的重要手段,包括人工自动免疫、人工被动免疫和过继免疫。 人工自动免疫是给机体接种疫苗或类毒素等抗原物质,刺激机体产生特异性免疫。国内常将用细菌制作的人工主动免疫的生物制品称为菌苗,而将用病毒、立克次体螺旋体等制成的生物制品称为疫苗,而国际上把细菌性制剂,病毒性制剂及类毒素统称为疫苗。经人工自动免疫产生的免疫力出现较慢,但免疫力较持久,故临床上多用于预防。人工自动免疫制剂其主要有灭活疫苗、减毒活疫苗、类毒素、以及各种新型疫苗。 人工被动免疫是给机体输入抗体等制剂,使机体获得特异性免疫力,输入抗体后立即获得免疫力,但维持时间短,约2~3周,临床上用于治疗或紧急预防。人工被动免疫的生物制品主要有抗毒素、抗菌血清与抗病毒血清、胎盘球蛋白和血浆丙种球蛋白。 过继免疫治疗是指给患者转输具有在体内继续扩增效应细胞的一种疗法。如给免疫缺陷病患者转输骨髓细胞;给肿瘤患者输入体外激活扩增的特异肿瘤浸润淋巴细胞或非特异性的LAK细胞等。应用时应考虑供者与受者之间HLA型别是否相同,否则输注的细胞会被迅速清除,或者发生移植物抗宿主反应。再如造血干细胞移植:取患者自身或异体骨髓或脐血输入患者,移植物中的多能干细胞可在体内定居、增殖、分化、使患者恢复造血功能和形成免疫力。造血干细胞移植可用于治疗再生障碍性贫血、白血病以及某些免疫缺陷病和自身免疫病等。 在医学制剂影响免疫功能的制剂主要有两类:免疫增强剂和免疫仰制剂。免疫增强剂是指通过不同方式,达到增强机体免疫力的一类免疫治疗药物。临床上常用于治疗与免疫功能低下有关的疾病及免疫缺陷病。免疫增强剂种类很多,按其作用的先决条件可分为三类:一是免疫替代剂,用来代替某些具有免疫增强作用的生物因子的药物。按其作用机制可分为提高巨噬细胞吞噬功能的药物,提高细胞免疫功能的药物,提高体液免疫功能的药物等;按其作用性质又可分为特异性免疫增强剂和非特异性免疫增强剂;按其来源则可分为细菌性免疫增强剂及非细菌性免疫增强剂。二是免疫恢复剂,能增强被抑制的免疫功能,但对正常免疫功能作用不大。常用的免疫增强剂如:卡介苗、短小棒状杆菌、内毒素、免疫核糖核酸、胸腺素、转移因子、双链聚核苷酸、佐剂等。免疫抑制剂是对机体的免疫反应具有抑制作用的药物。能抑制与免疫反应有关细胞的增殖和功能,能降低抗体免疫反应的制剂。常用的免疫抑制剂主要有五类:(1)糖皮质激素类,如可的松和强的松、泼尼松龙等;(2)微生物代谢产物,如环孢菌素和藤霉素等;(3)抗代谢物,如硫唑嘌呤和6-巯基嘌呤等;(4)多克隆和单克隆抗淋巴细胞抗体,如抗淋巴细胞球蛋白和OKT3等;(5)烷化剂类,如环磷酰胺等。 免疫学诊断是指应用免疫学原理和方法对传染病、免疫性疾病等进行和免疫功能进行测定。由于免疫学检测具有高特异性和敏感性,因此常用临床诊断的一种重要手段。目前常用的免疫诊断方法具有体液免疫试验。细胞免疫试验和皮肤试验三种。 抗原抗体反应在体内表现为溶细胞、杀菌、促进吞噬、中和毒素或引起免疫病理损伤等;在体外可出现凝集、沉淀、细胞溶解和补体结合等可见反应。由于抗体主要存在于血清中,临床上多用血清标本进行试验,故体外的抗原抗体反应曾被称为血清学反应。但随着免疫学

干细胞与组织工程重大项目课题申请指引

附件1: 863 计划生物和医药技术领域 “干细胞与组织工程”重大项目 课题申请指南 一、指南说明 “十一五”期间,本重大项目以替代、修复或再造人体各种组织器官的“再生医学”为主线,以干细胞与克隆技术、组织工程技术、组织器官代用品和再生医学相关评价体系为重点,利用多学科、多技术交叉合作的关键技术和资源平台,建立具有一定规模的高水平研究、生产和应用基地,形成我国再生医学工程研究开发技术体系,研制具有自主知识产权的系列干细胞与组织工程产品及代用品,建立、完善和细化相应的技术标准、准入规范和相关伦理学指导原则,培育和带动新兴产业,加快实现部分干细胞、组织工程产品和组织器官代用品的产业化,从而使我国再生医学研究和应用的整体水平进入世界先进行列,部分关键技术和产品达到国际领先水平。 本重大项目的任务分解及主要任务指标为:完成1-2 种重要退行性疾病的治疗性克隆临床前研究,力争获得第一个治疗性克隆的临床批 文;完成3-5 种重要疾病干细胞治疗的临床前研究,获得SFDA或卫生 行政主管部门的临床批文,力争1-2种治疗技术或产品 完成临床试验,获得相应证书;初步建立1-2 个国家级人类(疾病)胚胎干细胞库,建立干细胞库有关的硬件标准和管理规范;4-6 种具有自主知识产权的系列组织工程产品获得SFDA临床批文,力争 2-3 项完成临床试验并获得许可证;开发3-5 项具有自主知识产权的新型组织器官代用品,力争获得生产许可证;建立3-5 种针对干细胞和组织工程应用的灵长类动物疾病与评价模型;申请专利200 项左右,其中

10项左右为国际PCT专利,力争10%获得授权。 上述任务分解除个别保密课题外,所有课题均通过公开发布课题申请指南落实任务。 此次发布的是本重大项目课题申请指南,拟安排的主要内容包括干细胞与治疗性克隆、组织工程技术与产品研制、组织器官代用品研发及灵长类动物疾病与评价模型等,拟支持的经费为 2 亿元人民币。课题支持强度依据所承担任务和完成指标而定,课题支持年限为5 年,但依据中期评估进行滚动支持。 二、指南内容 课题 1 、帕金森病或卢迦雷氏病的治疗性克隆研究与应用研究目标:完成治疗性克隆技术及治疗性克隆干细胞的临床前研究及有效性和安全性评价,向SFDA申报临床批文,争取在治疗性克隆领域有所突破。此课题是重大项目总体目标中的重要组成部分和亮点,也是体现本项目水平的重要指标之一。 主要研究内容及考核指标:针对帕金森病或卢迦雷氏病等神经系统退行性疾病,建立治疗性克隆胚胎干细胞系,建立治疗性克隆临床应用评价体系和相关检测手段,完成治疗性克隆技术及治疗性克隆干细胞的有效性和安全性评价等临床前研究,申报SFDA临床批文,争取率先将1-2 种治疗性克隆技术应用到临床。申请30 项左右国家专利及部分PCT力争10%获得授权。 课题支持年限:五年。 课题经费来源及构成:课题拟支持的国拨总经费为1500 万元人民币。作为战略高技术类课题,以国拨经费为主,课题申请单位配套相应的人员经费。 课题2、我国人类(疾病)胚胎干细胞库的建立与应用 研究目标:有计划、有组织、有目标、有分工地利用好、保护好我

驳复仇议阅读题及答案.doc

驳复仇议阅读题及答案 《驳复仇议》出自于唐代文学家柳宗元的一篇驳论性的奏议,这篇奏议阐述了"调"即"和谐"在处理社会矛盾中的重要作用。以下是我给你推荐的驳复仇议阅读题及参考答案,希望对你有帮助! 《驳复仇议》阅读原文 臣伏见天后①时,有同州下邦人徐元庆者,父爽为县吏赵师韫所杀,卒能手刃父仇,束身归罪。当时谏臣陈予昂建议诛之而旌其闾,且请编之于令,永为国典。臣窃独过之。 臣闻礼之大本,以防乱也。若日无为贼虐,凡为子者杀无赦。刑之大本,亦以防乱也。若曰无为贼虐,凡为理者杀无赦。其本则合,其用则异,旌与诛莫得而并焉。诛其可旌,兹谓滥;黩刑甚矣。旌其可诛,兹谓僭;坏礼甚矣。果以是示于天下,传于后代,趋义者不知所向,违害者不知所立,以是为典,可乎? 盖圣人之制,穷理以定赏罚,本情以正褒贬,统于一而已矣。向使刺谳②其诚伪,考正其曲直,原始而求其端,则刑礼之用,判然离矣。何者?若元庆之父,不陷于公罪,师韫之诛,独以其私怨,奋其吏气,虐于非辜,州牧不知罪,刑官不知问,上下蒙冒,吁号不闻;而元庆能以戴天为大耻,枕戈为得礼,处心积虑,以冲仇人之胸,介然自克,即死无憾,是守礼而行义也。执事者宜有惭色,将谢之不暇,而又何诛焉? 其或元庆之父,不

免于罪,师韫之诛,不愆于法,是非死于吏也,是死于法也。法其可仇乎?仇天子之法,而戕奉法之吏,是悖骜而凌上也。执而诛之,所以正邦典,而又何旌焉? 且其议曰:人必有子,子必有亲,亲亲相仇,其乱谁救?是惑于礼也甚矣。礼之所谓仇者,盖其冤抑沉痛,而号无告也;非谓抵罪触法,陷于大戮。而曰彼杀之,我乃杀之,不议曲直,暴寡胁弱而已。其非经背圣,不亦甚哉!《周礼》:调人,掌司万人之仇。凡杀人而义者,令勿仇;仇之则死。有反杀者,邦国交仇之。又安得亲亲相仇也?《春秋公羊传》曰:父不受诛,子复仇可也。父受诛,子复仇,此推刃③之道,复仇不除害。今若取此以断两下相杀,则合于礼矣。 且夫不忘仇,孝也;不爱死,义也。元庆能不越于礼,服孝死义,是必达理而闻道者也。夫达理闻道之人,岂其以王法为敌仇者哉?议者反以为戮,黩刑坏礼,其不可以为典,明矣。 请下臣议附于令,有断斯狱者,不宜以前议从事。谨议。 【注】①天后:武则天。②刺谳:刺,探寻;谳,议罪。 ③推刃:往来项杀。 《驳复仇议》阅读题目 4.对下列句子中加点的词的解释,不正确的一项是(3分) A.臣窃独过之过:认为不对 B.旌其可诛,兹谓僭僭:僭越 C.不愆于法愆:罪过

驳《复仇议》阅读答案附翻译

驳《复仇议》阅读答案附翻译 驳《复仇议》 [唐]柳宗元 臣伏见天后①时,有同州下邦人徐元庆者,父爽为县吏赵师韫所杀,卒能手刃父仇,束身归罪。当时谏臣陈予昂建议诛之而旌其闾,且请编之于令,永为国典。臣窃独过之。 臣闻礼之大本,以防乱也。若日无为贼虐,凡为子者杀无赦。刑之大本,亦以防乱也。若曰无为贼虐,凡为理者杀无赦。其本则合,其用则异,旌与诛莫得而并焉。诛其可旌,兹谓滥;黩刑甚矣。旌其可诛,兹谓僭;坏礼甚矣。果以是示于天下,传于后代,趋义者不知所向,违害者不知所立,以是为典,可乎? 盖圣人之制,穷理以定赏罚,本情以正褒贬,统于一而已矣。向使刺谳②其诚伪,考正其曲直,原始而求其端,则刑礼之用,判然离矣。何者?若元庆之父,不陷于公罪,师韫之诛,独以其私怨,奋其吏气,虐于非辜,州牧不知罪,刑官不知问,上下蒙冒,吁号不闻;而元庆能以戴天为大耻,枕戈为得礼,处心积虑,以冲仇人之胸,介然自克,即死无憾,是守礼而行义也。执事者宜有惭色,将谢之不暇,而又何诛焉? 其或元庆之父,不免于罪,师韫之诛,不愆于法,是非死于吏也,是死于法也。法其可仇乎?仇天子之法,而戕奉法之吏,是悖骜而凌上也。执而诛之,所以正邦典,而又何旌焉? 且其议曰:人必有子,子必有亲,亲亲相仇,其乱谁救?是惑于礼也甚矣。礼之所谓仇者,盖其冤抑沉痛,而号无告也;非谓抵罪触法,陷于大戮。而曰彼杀之,我乃杀之,不议曲直,暴寡胁弱而已。其非经背圣,不亦甚哉!《周礼》:调人,掌司万人之仇。凡杀人而义者,令勿仇;仇之则死。有反杀者,邦国交仇之。又安得亲亲相仇也?《春秋公羊传》曰:父不受诛,子复仇可也。父受诛,子复仇,此推刃③之道,复仇不除害。今若取此以断两下相杀,则合于礼矣。 且夫不忘仇,孝也;不爱死,义也。元庆能不越于礼,服孝死义,是必达理而闻道者也。夫达理闻道之人,岂其以王法为敌仇者哉?议者反以为戮,黩刑坏礼,其不可以为典,明矣。 请下臣议附于令,有断斯狱者,不宜以前议从事。谨议。 【注】①天后:武则天。②刺谳:刺,探寻;谳,议罪。③推刃:往来项杀。 4.对下列句子中加点的词的解释,不正确的一项是 A.臣窃独过之过:认为不对 B.旌其可诛,兹谓僭僭:僭越 C.不愆于法愆:罪过 D.不宜以前议从事从事:处置 5.下列各组句子中,加点词的意义和用法相同的一组是 A.旌与诛莫得而并焉臣与将军戮力而攻秦。 B.而又何诛焉王问:何以知之? C.我乃杀之今其智乃反不能及 D.是必达理而闻道者也虽一龙发机,而七首不动。 6.下列对原文有关内容的概括和分析,不正确的一项是 A.徐元庆杀了父亲的仇人后投案认罪。陈于昂建议,先处死徐元庆,再在他的家乡表彰他,并把这个案例编入法律文书中。作者认为,陈子昂的建议是错误的。 B.作者认为,礼与刑根本作用一致,但在实际运用中有区别,不能对同一个人既施死刑又行褒奖。自相矛盾的做法,公之于众,只会让天下人无所适从。

常用免疫学检验技术的基本原理

常用免疫学检验技术的基本原理 免疫学检测即是根据抗原、抗体反应的原理,利用已知的抗原检测未知的抗体或利用已知的抗体检测未知的抗原。由于外源性和内源性抗原均可通过不同的抗原递呈途径诱导生物机体的免疫应答,在生物体内产生特异性和非特异性T 细胞的克隆扩增,并分泌特异性的免疫球蛋白(抗体)。由于抗体-抗原的结合具有特异性和专一性的特点,这种检测可以定性、定位和定量地检测某一特异的蛋白(抗原或抗体)。免疫学检测技术的用途非常广泛,它们可用于各种疾病的诊断、疗效评价及发病机制的研究。 最初的免疫检测方法是将抗原或抗体的一方或双方在某种介质中进行扩散,通过观察抗原-抗体相遇时产生的沉淀反应,检测抗原或抗体,最终达到诊断的目的。这种扩散可以是蛋白的自然扩散,例如环状沉淀试验、单向免疫扩散试验、双向免疫扩散实验。单向免疫扩散试验就是在凝胶中混入抗体,制成含有抗体的凝胶板,而将抗原加入凝胶板预先打好的小孔内,让抗原从小孔向四周的凝胶自然扩散,当一定浓度的抗原和凝胶中的抗体相遇时便能形成免疫复合物,出现以小孔为中心的圆形沉淀圈,沉淀圈的直径与加入的抗原浓度成正比。 利用蛋白在不同酸碱度下带不同电荷的特性,可以利用人为的电场将抗原、抗体扩散,例如免疫电泳试验和双向免疫电泳。免疫电泳首先将抗原加入凝胶中电泳,将抗原各成分依次分散开。然后沿电泳方向平行挖一直线形槽,于槽内加入含有针对各种抗原的混合抗体,让各抗原成分与相应抗体进行自然扩散,形成沉淀线。然后利用标准的抗原-抗体沉淀线进行抗原蛋白(或抗体)的鉴别。上述的方法都是利用肉眼观察抗原-抗体反应产生的沉淀,因此灵敏度有很大的局限。比浊法引入沉淀检测产生的免疫比浊法就是利用浊度计测量液体中抗原-抗体反应产生的浊度,根据标准曲线来计算抗原(或抗体)的含量。该方法不但大大提高了检测的灵敏度,且可对抗原、抗体进行定量的检测。

免疫学发展简史

免疫学发展简史 分三个时期:①经验免疫学时期(公元前400年~18世纪末); ②免疫学科建立时期(19世纪~1975年);③现代免疫学时期(1975年至今)。 一、经验免疫学时期(公元前400年~18世纪末) (一)天花的危害 天花是一种古老的、世界流行的烈性传染病,死亡率可高达25%~40%,我国民间早有“生了孩子算一半,得了天花才算全”的说法。患天花痊愈后留下永久的疤痕,但可获得终身免疫。 16世纪由于西班牙殖民者侵略,将天花传播到美洲,墨西哥土著人从16世纪初(1518年)的2000~3000万人到16世纪末减少到100万人,阿茨特克帝国消亡。16世纪中期之后向南进发,在美洲中部毁灭了玛雅和印加文明,随后又毁灭了秘鲁。 (二)人痘苗接种 1.人痘苗接种实践: 中医称天花为“痘疮”,据史书记载人痘苗接种预防天花的方法是在公元前约400年由我们中华民族的祖先建立的。Zinsser微生物学(1988):发明于中国2000多年之前。 明庆隆年间(1567~1572);16~17世纪人痘苗接种预防天花已在全国普遍展开。清康熙27年(1688)俄国曾派医生到北京学习种痘技术。并经丝绸之路东传至朝鲜、日本和东南亚国家,西传至欧亚、北非及北美各国。 1700年传入英国/Momtagu夫人在英国积极推广人痘苗接种中起了重要的作用。 1721~1722年天花在英国爆发流行期间,英国皇家学会在国王的特许下,主持进行了用犯人和孤儿做人痘苗接种的试验,均获得了成功,试验者无一人死于天花。在此基础上,1722年给英国威尔士王子的两个女儿(一个9岁,一个11岁)也进行了人痘苗接种,也都获得成功。 2.人痘苗接种意义:有三个方面: ①能有效预防天花。 ②在接种方法、痘苗的制备和保存建立了一整套完整的科学方法,为以后疫苗的发展提供了丰富的经验和借鉴。 清代吴谦所著的《医宗金鉴·幼科种痘心法要旨》(1742年)中介绍了四种接种法:痘衣法-痘浆法-旱苗法-水苗法。并指出这些方法的优劣:“水苗为上,旱苗次之,痘衣多不应验,痘浆太涉残忍。” 对痘苗保存指出:“若遇热则气泄,日久则气薄,触污秽则气不清,藏不洁则气不正,此蓄苗之法。”“须贮新磁瓶内,上以物密覆

大学语文重点句翻译练习

重点句翻译 一、《诗经》 1、王事靡盬,不遑启处;忧心孔疾,我行不来 2、昔我往矣,杨柳依依;今我来思,雨雪霏霏 二、《郑伯克段于鄢》 1、制,岩邑也,虢叔死焉,亻它邑唯命 2、都城过百雉,国之害也 3、姜氏何厌之有,不如早为之所,无使滋蔓,蔓,难图也 4、多行不义,必自毙,子姑待之 5、国不堪贰,君将若之何 6、厚将得众,不义不暱,厚将崩 7、大叔完聚,缮甲兵,具卒乘,将袭郑 8、公赐之食,食舍肉 9、君何患焉,若阙地及泉,隧而相见,其谁曰不然 10、孝子不匮,永锡尔类,君将不堪 11、今京不度,非制也,君将不堪 三、《燕昭王求士》 1、先趋而后患,先问而后嘿,则什己者至 2、此古服道致士之法也 3、燕国殷富,士卒乐佚轻战 4、燕兵独追北,人至临淄,尽取齐宝,烧其宫室宗庙 5、诎指而事之,北面而受学,则百己者至 6、今王诚欲致士,先从隗始,隗且见事,况贤于隗者乎,岂远千里哉 7、冯几据杖,眄视指使,则厮役之人至 四、《管晏列传》 1、仓廪实而知礼节,衣食足而知荣辱,上服度则六亲固 2、下令如流水之原,令顺民心 3、吾闻君子诎于不知己而信于知己者 4、通货积财,富国强兵,与俗同好恶 5、将顺其美,匡救其恶,故上下能相亲也 6、少时常与鲍叔牙游,鲍叔知其贤,管仲贫困,常欺鲍叔,鲍叔终善遇之,不以为言 7、吾尝三仕三见逐于君,鲍叔不以为我不肖,知我不遭时也 8、知我不羞小节而耻功名不显于天下也 1

9、知与之为政,政之宝也 10、其在朝,君语及之,即危言;语不及之,即危行 五、《苏武传》 1、武帝嘉其义,乃遣武以中郎将使持节送匈奴使留在汉者,因厚赂单于,答其善意 2、见犯乃死,重负国 3、复举剑拟之,武不动 4、空以身膏草野,谁复知之 5、武既至海上,廪食不至,掘野鼠去草实而食之 6、张胜闻之,恐前语发,以状语武 7、单于壮其节,朝夕遣人候问武,而收系张胜 8、凿地为坎,置煴火,覆武其上,蹈其背以出血 9、且单于信女,使决人死生,不平心持正,反欲斗两主,观祸败 10、天雨雪,武卧啮雪,与旃毛并咽之,数日不死 11、自分已死久矣,王必欲降武,请毕今日之欢,效死于前 12、收族陵家,为世大戮,陵尚复何顾乎 六、《先秦诸子语录》 1、其身正,不令而行;其身不正,虽令不从 2、老吾老以及人之老,幼吾幼以及人之幼,天下可运于掌 3、彼窃钩者诛,窃国者为诸侯,诸侯之门,而仁义存焉 七、《谏逐客书》 1、是以泰山不让土壤,故能成其大;河海不择细流,故能就其深;王者不却众庶,故能明其德 2、此所谓“藉寇兵而赍盗粮”者也 3、臣闻吏议逐客,窃以为过矣 4、今乃弃黔首以资敌国,却宾客以业诸侯 八、《驳复仇议》 1、其本则合,其用则异,旌与诛莫得而并焉 2、向使刺谳其诚伪,烤正其曲直,原始而求其端,则刑礼之用,判然离矣 3、元庆能不越于礼,服孝死义,是必达理而闻道者也 4、执事者宜有惭色,将谢之不暇,而又何诛焉 5、盖圣人之制,穷理以定赏罚,本情以正褒贬,终于一而已矣 6、仇天子之法,而戕奉法之吏,是悖骜而凌上也,是非死于吏也,是死于法也,法其可仇乎 九、《留侯论》

干细胞抗衰老行业现状分析资料

目录 一、干细胞行业的“万用功能“及发展前景 (1) 二、干细胞行业相关政策分析 (2) 三、干细胞抗衰老作用机理 (2) 1.骨髓间充质干细胞 (3) 2.脐带血干细胞 (4) 3.脂肪干细胞 (5) 4.胚胎干细胞 (5) 四、干细胞产业链分析 (6) 五、干细胞国内相关企业分析 (7) 1.中源协和:中国干细胞产业链的整合者,全面布局上下游 (7) 2.北科生物:干细胞技术全球领先,有望成为中国的“苹果” (8) 3.金卫医疗:通过CCBC股权间接经营干细胞存储业务 (9) 4.冠昊生物:依托技术优势进军干细胞治疗领域 (10)

图表目录 图表 1:干细胞治疗应用方向 (3) 图表 2:干细胞抗衰老行业相关政策 (4) 图表 3:随着年龄的增长,骨髓中干细胞数目急剧下降 (5) 图表 4:小鼠骨髓间充质干细胞具有抗衰老作用 (6) 图表 5:脐带血干细胞可以促进细胞增殖,修复受伤组织 (6) 图表 6:肌肉注射胚胎干细胞后各系统疗效(临床改善指数) (7) 图表 7:干细胞产业链 (8) 图表 8:北科生物发展历程 (11) 图表 9:金卫医疗发展历程 (12)

一、干细胞行业的“万用功能“及发展前景 干细胞是具有自我复制和多向分化潜能的原始细胞,是机体的起源细胞,是形成人体各种组织器官的原始细胞。在一定条件下,它可以分化成多种功能细胞或组织器官,医学界称其为“万用细胞”,对应干细胞治疗具有极大的潜力。干细胞按发育状态分为胚胎干细胞、成体干细胞。按分化潜能分为全能干细胞、多能干细胞、单能干细胞。 干细胞治疗则是把健康的干细胞移植到病人或自己体内,以达到修复病变细胞或重建功能正常的细胞和组织的目的。即干细胞治疗从细胞层上治疗疾病,相较很多传统治疗方法具有无可比拟的优点: (1)安全:低毒性(或无毒性); (2)在尚未完全了解疾病发病的确切机理前也可以应用; (3)治疗材料来源充足; (4)治疗范围广阔; (5)是最好的免疫治疗和基因治疗载体; (6)传统疗法认为是“不治之症”的疾病,又有了新的疗法和新的希望。 因此干细胞治疗作为一种比较优势突出的新型治疗,临床上应用的领域包括治疗心血管疾病、神经系统疾病、血液病、肝病、肾病、糖尿病、骨关节疾病等,现在比较成熟的如应用骨髓移植治疗白血病等恶性血液病,随着未来越来越多临床试验的成功,产业发展前景将十分广阔。 图表 1:干细胞治疗应用方向 资料来源:银联信

免疫学诊断技术的发展及趋势

免疫学诊断技术的发展及趋势 免疫学广泛应用于三大方面:①传染病预防:接种菌苗、疫苗,使机体主动产生免疫力.严重急性呼吸道综合征(SARS)及艾滋病,终将有赖于疫苗的发明(详见第二十五章)。②疾病治疗:包括肿瘤、慢性传染病及超敏性疾病,可用抗体、细胞因子、体外扩增的免疫细胞及治疗性抗原疫苗治疗。③免疫诊断:按抗原与抗体及T 细胞受体特异结合的原理;按抗原能活化特异的适应性免疫应答,发展起多种特异敏感的免疫学诊断方法,已广泛用于ABO血型定型,传染病诊断,妊娠确诊等等 天花曾是人类历史上的烈性传染病,是威胁人类的主要杀手之一。在欧洲,十七世纪中叶,患天花死亡者达30%。我国早在宋朝(十一世纪)已有吸入天花痂粉预防天花的传说。到明代,即公元十七世纪七十年代左右,则有正式记载接种“人痘”,预防天花。从经验观察,将沾有疱浆的患者的衣服给正常儿童穿戴,或将天花愈合后的局部痂皮磨碎成细粉,经鼻给正常儿童吸入,可预防天花。这些方法在北京地区较为流行,且经陆上丝绸之路西传至欧亚各国,经海上丝绸之路,东传至朝鲜、日本及东南亚国家。英国于1721年流行天花期间,曾以少数犯人试种人痘预防天花成功,但因当时英国学者的保守,未予推广。由于种“人痘”预防天花具有一定的危险性,使这一方法未能非常广泛地应用。然而, 其传播至世界各国,对人类寻求预防天花的方法有重要的影响。 公元十八世纪后叶,英国乡村医生Jenner 观察到牛患有牛痘,局部痘疹酷似人类天花,挤奶女工为患有牛痘的病牛挤奶,其手臂部亦得“牛痘”,但却不得天花。于是他意识到接种“牛痘”可预防天花。为证实这一设想,他将牛痘接种于一8 岁男孩手臂,两个月后,再接种从天花患者来源的痘液,只致局部手臂疱疹,未引起全身天花。他于1798年公布了他的论文,把接种牛痘“Vaccination”(拉丁语中,牛写为Vacca),即接种牛痘,预防天花。在Jenner年代,人们全然不知天花是由天花病毒感染所致,而他在实践观察中,总结发现的种牛痘预防天花,既安全、又有效,是一划时代的发明。接种牛痘在十九世纪初至中叶,在欧洲广泛推广。总之,在十九世纪以前,人们从经验得知接种人痘或牛痘,可获得免疫力,预防天花,但对病原体及获得免疫的道理却全然不知。 细胞免疫学的发展明确了T 及B 淋巴细胞经表面受体识别抗原分子,受体

干细胞技术及发展

干细胞技术及发展 摘要:干细胞技术是生物技术领域最具有发展前景和后劲的前沿技术,其已成为世界高新技术的新亮点,势将导致一场医学和生物学革命。本文主要介绍干细胞及干细胞的相关技术与发展方向。 关键词:干细胞干细胞技术发展方向 1 概述 干细胞技术,又称为再生医疗技术,是指通过对于干细胞进行分离、体外培养、定向诱导、甚至基因修饰等过程,在体外繁育出全新的、正常的甚至更年轻的细胞、组织或器官,并最终通过细胞组织或器官的移植实现对临床疾病的治疗。 干细胞技术最显著的作用就是:能再造一种全新的、正常的甚至更年轻的细胞、组织或器官。由此人们可以用自身或他人的干细胞和干细胞衍生组织、器官替代病变或衰老的组织、器官,并可以广泛涉及用于治疗传统医学方法难以医治的多种顽症,诸如白血病、早老性痴呆、帕金森氏病、糖尿病、中风和脊柱损伤等一系列目前尚不能治愈的疾病。从理论上说,应用干细胞技术能治疗各种疾病,且其较很多传统治疗方法具有无可比拟的优点。 2 干细胞技术 干细胞有诱人的临床应用前景,因此干细胞技术的研究开发自然成为当今世界生物医药领域的一个热点研究方向。干细胞技术涉及许多方面,包括干细胞的鉴别、分离纯化、分裂增殖、诱导分化、冷冻保存、解冻使用等。还包括干细胞培养液、干细胞再生医学生物材料、干细胞产品保存包装材料等相关技术。以下着重简述来自不同发育阶段和不同组织的干细胞的相关技术。 2.1 干细胞 在细胞的分化过程中,细胞往往由于高度分化而完全失去了再分裂的能力,最终衰老死亡。机体在发展适应过程中为了祢补这一不足,保留了一部分未分化的原始细胞即干细胞。一旦生理需要,这些干细胞可按照发育途径通过分裂而产生分化细胞。 2.1.1干细胞的特点 (1)干细胞本身不是处于分化途径的终端。 (2)干细胞能无限的增殖分裂。 (3)干细胞可连续分裂几代,也可在较长时间内处于静止状态。

当今免疫学的发展

当今免疫学的发展 免疫学是生命科学及医学领域中的前沿学科,涉及抗感染免疫、血液病、自身免疫病、移植免疫和肿瘤免疫等诸多范畴。该学科近二十年来与细胞生物学、分子生物学、分子遗传学以及生物化学相互渗透,发展迅猛。分子生物学、分子遗传学以及细胞生物学的发展促进了分子免疫学、免疫遗传学以及免疫生物学等新的分支学科的形成,使人们在分子水平上对免疫系统的结构与功能有了更加深刻的认识。生命科学中许多重大问题的发现、解决或应用都首先与免疫学研究的突破有关,免疫学基础理论研究的突破不断导致生命科学领域的革命。自1960年迄今共有13位免疫学家获得诺贝尔医学奖。本文仅就免疫学研究近年来的发展现状以及今后的发展趋势做简要的评述。 1 免疫学在分子水平上的深化与发展 分子免疫学近年来的突破性进展层出不穷。例如,发现天然免疫系统可通过特异性受体识别病原体共有的保守性分子特征(pattern),称此种受体为特征识别受体(pattern recognition receptor,PRR)。目前对PRR分子结构与信号转导途径正在深入研究中,并探讨天然免疫系统对获得性免疫应答类型导向作用的分子机制。此外,应用单克隆抗体及分子生物学技术发现了大量膜分子,被统一命名的白细胞膜表面分化抗原(CD分子)已有250个之多。再之,对免疫球蛋白分子、主要组织相容性(抗原)复合物(major histocompa-bility complex, MHC)分子、T细胞和NK 细胞识别受体、补体分子、细胞因子以及趋化因子等的分子结构、生物学功能、基因结构等均有了相当深入的了解。近年来对淋巴细胞发育的分子机制研究也有突破性进展。例如,发现PU.1/Ikaros可调控T、B细胞的发育,GATA-3影响T谱系的发育,EIA/EBF/Pax可调控B细胞的发育等。对T细胞在胸腺内分化发育分子机制的研究表明,胸腺细胞膜分子、pTA/TCR分子、Bortch分子、CD30/CD153以及CD69等分子与其分化相关。此外还发现Ras-MAPK信号转导与阳性选择相关,而与阴性选择无关。对T、B细胞活化、增殖、分化、凋亡的分子机制研究发现,T、B细胞在免疫应答过程中涉及多种膜受体分子如TCR/BCR复合分子、粘附分子、趋化因子受体、补体受体、Fc受体以及Fas等。这些分子在免疫应答过程中发挥不同的作用。小鼠和人辅助性T细胞(Th)可在不同的环境条件下(如APC类型、抗原种类、细胞因子)发育分化为不同功能性Th细胞(如Th1、Th2、Th3等)亚群。目前区分这些T细胞亚群的主要指标是它们分泌细胞因子的特点,可以预测在不久的将来还会发现可用于区别不同Th细胞亚群的膜表面分子。Th1/Th2平衡调节可导向免疫应答类型,其在免疫性疾病中的作用倍受重视。 2 免疫系统与神经内分泌系统的相互作用 80年代初发现免疫细胞可以合成阿片肽,其后对免疫系统和神经内分泌系统相互关系的研究进展迅速,极大地丰富了我们对机体内环境稳定机制的理解。目前已证实免疫细胞表面具有神经递质和内分泌激素受体,而神经细胞表面也具有细胞因子受体,因此各系统可通过各自表面受体及其释放的介质进行信息交流及功能调节,藉以维持机体内环境的稳定。

免疫学发展简史

免疫学发展简史 分三个时期:①经验免疫学时期(公元前400年~18世纪末);②免疫学科建立时期(19世纪~1975年);③现代免疫学时期(1975年至今)、 一、经验免疫学时期(公元前400年~18世纪末) (一)天花得危害 天花就是一种古老得、世界流行得烈性传染病,死亡率可高达25%~40%,我国民间早有“生了孩子算一半,得了天花才算全”得说法。患天花痊愈后留下永久得疤痕,但可获得终身免疫。 16世纪由于西班牙殖民者侵略,将天花传播到美洲,墨西哥土著人从16世纪初(1518年)得2000~3000万人到16世纪末减少到100万人,阿茨特克帝国消亡、16世纪中期之后向南进发,在美洲中部毁灭了玛雅与印加文明,随后又毁灭了秘鲁。 (二)人痘苗接种 1、人痘苗接种实践: 中医称天花为“痘疮",据史书记载人痘苗接种预防天花得方法就是在公元前约400年由我们中华民族得祖先建立得、Zinsser微生物学(1988):发明于中国2000多年之前、 明庆隆年间(1567~1572);16~17世纪人痘苗接种预防天花已在全国普遍展开。清康熙27年(1688)俄国曾派医生到北京学习种痘技术、并经丝绸之路东传至朝鲜、日本与东南亚国家,西传至欧亚、北非及北美各国。 1700年传入英国/Momtagu夫人在英国积极推广人痘苗接种中起了重要得作用。 1721~1722年天花在英国爆发流行期间,英国皇家学会在国王得特许下,主持进行了用犯人与孤儿做人痘苗接种得试验,均获得了成功,试验者无一人死于天花、在此基础上,1722年给英国威尔士王子得两个女儿(一个9岁,一个11岁)也进行了人痘苗接种,也都获得成功。 2。人痘苗接种意义:有三个方面: ①能有效预防天花。 ②在接种方法、痘苗得制备与保存建立了一整套完整得科学方法,为以后疫苗得发展提供了丰富得经验与借鉴。 清代吴谦所著得《医宗金鉴·幼科种痘心法要旨》(1742年)中介绍了四种接种法:痘衣法—痘浆法-旱苗法-水苗法。并指出这些方法得优劣:“水苗为上,旱苗次之,痘衣多不应验,痘浆太涉残忍、” 对痘苗保存指出:“若遇热则气泄,日久则气薄,触污秽则气不清,藏不洁则气不正,此蓄苗之法、”“须贮新磁瓶内,上以物密覆之,置之洁净之所,清凉之处。” 痘苗有“时苗"与“熟苗”之分,开始采用得痘痂叫时苗,经人体接种传代后制备得叫熟苗。清代朱奕梁编著得《种痘心法》中写道:“其苗传种愈久,则药力之

柳宗元驳复仇议翻译

驳复仇议 臣伏见天后时①,有同州下邽人徐元庆者②,父爽为县尉赵师韫所杀③,卒能手刃父仇,束身归罪④。当时谏臣陈子昂建议诛之而旌其闾⑤,且请“编之于令,永为国典。”臣窃独过之⑥。 注释: ①伏见:旧时臣下对君主有所陈述时的表敬之辞,可译为知道,了解。天后:即武则天(624-705),名武瞾。690年,废睿宗李旦自立为皇帝,改洛阳为神都,建立武周王朝,在位十六年。705年,武则天病笃,宰相张柬之发动兵变,迫使武氏退位,史称神龙革命。中宗李哲复位,恢复唐朝。 ②同州:唐代州名,今陕西渭南市大荔县一带地区。下邽(guī):县名,今陕西省渭南县。徐元庆:当时某驿馆的服务人员,徐元庆替父报仇,谋杀官员赵师蕴案是武则天时轰动一时的谋杀案。 ③县尉:县令的属官,专司当地的治安工作。或称御史大夫。 ④卒:最后,最终。束身归罪:自首。 ⑤陈子昂:(661—702),字伯玉。武后时曾任右拾遗,为谏诤之官。旌(j īng):表彰。闾:里巷的大门。 ⑥过:错误,失当。 译文:微臣知道则天皇后时,同州下邽县有个叫徐元庆的人,他的父亲徐爽被县尉赵师韫杀害,他最后能亲手杀掉他父亲的仇人,并且自己捆绑着身体到官府自首。当时的谏官陈子昂建议将他处以死罪,同时在他的家乡表彰他的行为,并请朝廷将这种处理方式“编入法令,永远作为国家的法律制度”。臣私下认为,这样做是不对的。 臣闻礼之大本,以防乱也。若曰无为贼虐①,凡为子者杀无赦。刑之大本,亦以防乱也,若曰无为贼虐,凡为治者杀无赦。其本则合,其用则异。旌与诛莫得而并焉。诛其可旌,兹谓滥,黩②刑甚矣。旌其可诛,兹谓僣③,坏礼甚矣。果以是示于天下,传于后代,趋义者不知所向,违害者不知所立,以是为典可乎盖圣人之制,穷理以定赏罚,本情以正褒贬,统于一而已矣。 注释: ①贼虐:残害,践踏 ②黩(dú)刑:滥用刑法。黩,轻率。 ③僭(jiàn):越过,超出本分。

相关文档
相关文档 最新文档