Sertoli cell: Difference between revisions

From Embryology
mNo edit summary
mNo edit summary
 
(7 intermediate revisions by the same user not shown)
Line 13: Line 13:


{|
{|
| [[File:Enrico Sertoli.jpg|150px|alt=Enrico Sertoli (1842 - 1910)]]
| [[File:Enrico Sertoli.jpg|100px|alt=Enrico Sertoli (1842 - 1910)]]
Enrico Sertoli (1842 - 1910)
Enrico Sertoli (1842 - 1910)
| Sertoli cell are named after Enrico Sertoli (1842 - 1910), an Italian physiologist and histologist.  
| Sertoli cell are named after Enrico Sertoli (1842 - 1910), an Italian physiologist and histologist.  
Line 48: Line 48:
==Development Overview==
==Development Overview==
[[File:Mouse- gonadal supporting cell development.jpg|thumb|alt=Gonadal supporting cell development|Gonadal supporting cell development]]
[[File:Mouse- gonadal supporting cell development.jpg|thumb|alt=Gonadal supporting cell development|Gonadal supporting cell development]]
Ultrastructural description of human Sertoli cells{{#pmid:11732573|PMID11732573}}
* '''7 weeks'''  - first morphologically recognised in testicular cords, organised as primordial germ cells surrounded by pre-Sertoli cells.
* '''7 to 8 weeks''' - basal lamina of the cords becomes distinguishable, pre-Sertoli cells the rough endoplasmic reticulum develops.
* '''14 to 20 weeks''' - pre-Sertoli cells maintain their general morphology whereas the most significant change is the maximum development of Leydig cells.
{{Seminiferous Tubule puberty table}}
{{Seminiferous Tubule puberty table}}


Line 67: Line 72:
(see also review{{#pmid:7711190|PMID7711190}})
(see also review{{#pmid:7711190|PMID7711190}})


Ultrastructural description of human Sertoli cells{{#pmid:11732573|PMID11732573}}
* '''7 weeks'''  - first morphologically recognised in testicular cords, organised as primordial germ cells surrounded by pre-Sertoli cells.
* '''7 to 8 weeks''' - basal lamina of the cords becomes distinguishable, pre-Sertoli cells the rough endoplasmic reticulum develops.
* '''14 to 20 weeks''' - pre-Sertoli cells maintain their general morphology whereas the most significant change is the maximum development of Leydig cells.


Molecular factors:  
Molecular factors:  
Line 79: Line 80:


:'''Links:'''[[Sertoli cell]]
:'''Links:'''[[Sertoli cell]]
<div id="Leydig cell"></div>


==Blood-Testis Barrier ==
==Blood-Testis Barrier ==


Within the testis seminiferous tubules the Sertoli cells located near the basement membrane act as an initial cellular barrier with many functions, but often described as forming a "blood-testis barrier". (see review{{#pmid:21134990|PMID21134990}}
Within the testis seminiferous tubules the Sertoli cells located near the basement membrane act as an initial cellular barrier with many functions, but often described as forming a "blood-testis barrier". (see reviews{{#pmid:22039149|PMID22039149}}{{#pmid:21134990|PMID21134990}} The BTB physically divides the seminiferous epithelium into two compartments: the basal and apical (adluminal).


Functions:
Functions:
Line 90: Line 89:
* establish a basal and adluminal (apical) compartment (specialized microenvironment)
* establish a basal and adluminal (apical) compartment (specialized microenvironment)
* provide an immunological privilege status of the testis (anti-sperm antibodies are not developed)
* provide an immunological privilege status of the testis (anti-sperm antibodies are not developed)
{|
|+ '''Rat Blood–Testis Barrier'''
|-
| colspan=2|A model depicting the migration of developing preleptotene spermatocytes across the blood–testis barrier (BTB}.{{#pmid:17698604|PMID17698604}}
|-
| [[File:Rat blood–testis barrier 02.jpg|400px]]
| [[File:Rat blood–testis barrier 03.jpg|400px]]
|-
| Movement of developing germ cells in the seminiferous epithelium of the adult testis.
Germ cells first have to break through the tight junctions at the BTB with minimal disruptions. After the opening of the BTB, the progression of germ cells relies solely on series of transient adherens junctions at the Sertoli–germ cell interface. Assembly and disassembly of junctional complexes at the basal and apical ectoplasmic specialization occurs continuously.
| When germ cells move along Sertoli cells, the original JAM/CAR/nectin trans-homodimers between Sertoli cells are replaced by the Sertoli–germ cell junctions composed of CAR–CAR/CAR–JAM-C/nectin-2–nectin-3/necl-5(PVR)–necl-2/JAM-B–JAM-C complexes plausibly through competitive binding. It is noted that CAR–CAR adhesion has not been directly demonstrated for Sertoli cell–germ cell interaction, but remains an attractive possibility.
|-
| colspan=2|Text from original figure legend.
|}
==Hormones==
These cells secrete several hormones, anti-Müllerian hormone ({{AMH}}), inhibin and activin.
Inhibin and activin provide positive and negative feedback respectively on follicle-stimulating hormone ({{FSH}}) secretion from the {{pituitary}}.
* activins - dimers of beta-A and/or beta-B subunits encoded by the genes INHBA and INHBB
* Follicle-stimulating hormone (FSH)-releasing protein (FRP) subunit is identical in structure to the beta-A subunit of inhibin.
===Anti-Mullerian Hormone===
Anti-Mullerian Hormone (Anti-Müllerian Hormone, {{AMH}}) is a glycoprotein that is produced by immature Sertoli cells.
Anti-Mullerian hormone (AMH) is a glycoprotein belonging to the transforming growth factor-beta (TGF-beta) family. During development, immature Sertoli cells produce AMH{{#pmid:1292986|PMID1292986}} to aid regression of Müllerian (paramesonephric) ducts in male sex developement.{{#pmid:28851672|PMID28851672}}
Follicle-stimulating hormone (FSH) promotes AMH transcription in the absence of androgen signaling, while {{Testosterone}} has been shown to inhibit the transcriptional activation of AMH.
:'''Links:''' {{Anti-Mullerian Hormone}}


==Abnormalities==
==Abnormalities==
Line 185: Line 220:


{{#pmid:19339813}}
{{#pmid:19339813}}
{{#pmid:22039149}}
===Articles===
===Articles===
{{#pmid:21147849}}
{{#pmid:21147849}}
Line 197: Line 234:


===Books===
===Books===
{{#pmid:25905331}}
{{#pmid:25905331}}
Skinner M. and Griswold M. [https://www.elsevier.com/books/sertoli-cell-biology/skinner/978-0-12-647751-1 '''Sertoli Cell Biology'''] (2004) Academic Press


===Search PubMed===
===Search PubMed===

Latest revision as of 22:53, 2 May 2019

Embryology - 16 Apr 2024    Facebook link Pinterest link Twitter link  Expand to Translate  
Google Translate - select your language from the list shown below (this will open a new external page)

العربية | català | 中文 | 中國傳統的 | français | Deutsche | עִברִית | हिंदी | bahasa Indonesia | italiano | 日本語 | 한국어 | မြန်မာ | Pilipino | Polskie | português | ਪੰਜਾਬੀ ਦੇ | Română | русский | Español | Swahili | Svensk | ไทย | Türkçe | اردو | ייִדיש | Tiếng Việt    These external translations are automated and may not be accurate. (More? About Translations)

Introduction

Adult Seminiferous tubule showing spermatozoa developmental stages
Adult Seminiferous tubule showing Sertoli cells and spermatozoa development

The sertoli cells are the first cells to be differentiated in development by SRY expression. Post-puberty these are the "support" cells for spermatozoa development and transport from the periphery to lumen of the seminiferous tubule. Sertoli cells form a barrier with cell junctions at the Sertoli cell-cell and Sertoli-germ cell interface.

The initial difference in male and female gonad development are dependent on testis-determining factor (TDF) the protein product of the Y chromosome SRY gene expression in the developing Sertoli cell. Recent studies have indicated that additional factors may also be required for full sex differentiation. The seminiferous tubules are considered the parenchyma of the testis. Within the developing testis the three main differentiating cell types are: gamete forming cells (spermatogonia), support cells (Sertoli cell) and hormone secreting cells (Leydig cell or interstitial cell).

Sertoli cell postnatal proliferation may be regulated by thyroid status. An animal model study of postnatal transient hypothyroidism has demonstrated Sertoli cell proliferation (6 to 8 fold increase) 2 days after the diet switch and remained elevated the next days.[1]


Genital Links: genital | Lecture - Medicine | Lecture - Science | Lecture Movie | Medicine - Practical | primordial germ cell | meiosis | endocrine gonad‎ | Genital Movies | genital abnormalities | Assisted Reproductive Technology | puberty | Category:Genital
Female | X | X inactivation | ovary | corpus luteum | oocyte | uterus | vagina | reproductive cycles | menstrual cycle | Category:Female
Male | Y | SRY | testis | spermatozoa | ductus deferens | penis | prostate | Category:Male
Historic Embryology - Genital 
General: 1901 Urinogenital Tract | 1902 The Uro-Genital System | 1904 Ovary and Testis | 1912 Urinogenital Organ Development | 1914 External Genitalia | 1921 Urogenital Development | 1921 External Genital | 1942 Sex Cords | 1953 Germ Cells | Historic Embryology Papers | Historic Disclaimer
Female: 1904 Ovary and Testis | 1904 Hymen | 1912 Urinogenital Organ Development | 1914 External Genitalia | 1914 Female | 1921 External Genital | 1927 Female Foetus 15 cm | 1927 Vagina | 1932 Postnatal Ovary
Male: 1887-88 Testis | 1904 Ovary and Testis | 1904 Leydig Cells | 1906 Testis vascular | 1909 Prostate | 1912 Prostate | 1914 External Genitalia | 1915 Cowper’s and Bartholin’s Glands | 1920 Wolffian tubules | 1935 Prepuce | 1935 Wolffian Duct | 1942 Sex Cords | 1943 Testes Descent | Historic Embryology Papers | Historic Disclaimer


Enrico Sertoli (1842 - 1910)

Enrico Sertoli (1842 - 1910)

Sertoli cell are named after Enrico Sertoli (1842 - 1910), an Italian physiologist and histologist.

His historic paper "The structure of seminiferous tubules and the development of (spermatids) in rats" have recently been translated from the original Italian into English.[2]

See also a biography written in 2002.[3]

Some Recent Findings

Historic testis drawing
  • Sertoli Cell Wt1 Regulates Peritubular Myoid Cell and Fetal Leydig Cell Differentiation during Fetal Testis Development[4] "Sertoli cells play a significant role in regulating fetal testis compartmentalization to generate testis cords and interstitium during development. The Sertoli cell Wilms' tumor 1 (Wt1) gene, which encodes ~24 zinc finger-containing transcription factors, is known to play a crucial role in fetal testis cord assembly and maintenance. ... In summary, Wt1 regulates the development of FLC and interstitial progenitor cell lineages through Notch signaling, and it also plays a role in PMC development. Collectively, these effects confer fetal testis compartmentalization. See also - Sertoli cells control peritubular myoid cell fate and support adult Leydig cell development in the prepubertal testis.[5]
  • Gadd45g is essential for primary sex determination, male fertility and testis development[6] "In humans and most mammals, differentiation of the embryonic gonad into ovaries or testes is controlled by the Y-linked gene SRY. Here we show a role for the Gadd45g protein in this primary sex differentiation. ...The molecular cause of the sex reversal was the failure of Gadd45g(-/-) XY gonads to achieve the SRY expression threshold necessary for testes differentiation, resulting in ovary and Müllerian duct development. These results identify Gadd45g as a candidate gene for male infertility and 46,XY sex reversal in humans."
More recent papers  
Mark Hill.jpg
PubMed logo.gif

This table allows an automated computer search of the external PubMed database using the listed "Search term" text link.

  • This search now requires a manual link as the original PubMed extension has been disabled.
  • The displayed list of references do not reflect any editorial selection of material based on content or relevance.
  • References also appear on this list based upon the date of the actual page viewing.


References listed on the rest of the content page and the associated discussion page (listed under the publication year sub-headings) do include some editorial selection based upon both relevance and availability.

More? References | Discussion Page | Journal Searches | 2019 References | 2020 References

Search term: Sertoli cell development | Sertoli cell barrier

Older papers  
These papers originally appeared in the Some Recent Findings table, but as that list grew in length have now been shuffled down to this collapsible table.

See also the Discussion Page for other references listed by year and References on this current page.

  • FGF signaling directs a center-to-pole expansion of tubulogenesis in mouse testis differentiation.[7] "These observations imply that center-to-pole FGF9 diffusion directs a poleward expansion of testiculogenic programs along the anteroposterior axis of developing XY gonads."

Development Overview

Gonadal supporting cell development
Gonadal supporting cell development

Ultrastructural description of human Sertoli cells[8]

  • 7 weeks - first morphologically recognised in testicular cords, organised as primordial germ cells surrounded by pre-Sertoli cells.
  • 7 to 8 weeks - basal lamina of the cords becomes distinguishable, pre-Sertoli cells the rough endoplasmic reticulum develops.
  • 14 to 20 weeks - pre-Sertoli cells maintain their general morphology whereas the most significant change is the maximum development of Leydig cells.
Testis - Seminiferous Tubule
Pre-puberty Post-puberty
Testis histology 006.jpg Testis histology 011.jpg
Cross-sectional view of the seminiferous tubule histology before and after puberty.

The sertoli cell cells provide support for spermatozoa development within the seminiferous tubule. In the mouse, these cells are derived from the coelomic epithelium along with other testis somatic cells.[9] Their differentiation is regulated by the presence of a Y chromosome and in turn regulates Leydig cell differentiation. Sertoli cells direct testis morphogenesis, organizing testis cord formation, establishing testis vasculature and inducing differentiation of peritubular myoid cells and fetal Leydig cells. At puberty the immature Sertoli cells cease to proliferate and differentiate. Activin A acts upon Sertoli cells to promote their embryonic proliferation[10]

Sertoli cells express the androgen receptor and receptors for follicle stimulating hormone (FSH).


Sertoli cell functions include:

  • regulation of spermatogenesis through endocrine FSH and testosterone
  • regulation of the intratubular and intercellular environment adluminal to the tight junctional complexes
    • meiotic and post-meiotic germ cells are sequestered by Sertoli-Sertoli junctional complexes
  • generate adluminal compartment isolated from both serum and lymph
  • attachment of germ cells through unique intermediate filament (desmosome-like junctions) and microfilament (actin-ectoplasmic specializations, ESs) junctions[11]
    • to prevent premature sloughing of immature germ cells from the seminiferous epithelium
    • desmosome-like junctions are initially present (up to step 8 spermatids)
    • ectoplasmic specializations then replace this junction (in step 8 spermatids)

(see also review[12])


Molecular factors:

  • Follicle Stimulating Hormone (FSH) -> Krüppel-like factor 4 (KLF4)
  • Krüppel-like factor 4 (KLF4) - zinc finger transcription factor, terminal differentiation of epithelial cells.
  • Epidermal Growth Factor (EGF)
  • Transforming Growth Factor-beta (TGFbeta)
Links:Sertoli cell

Blood-Testis Barrier

Within the testis seminiferous tubules the Sertoli cells located near the basement membrane act as an initial cellular barrier with many functions, but often described as forming a "blood-testis barrier". (see reviews[13][14] The BTB physically divides the seminiferous epithelium into two compartments: the basal and apical (adluminal).

Functions:

  • prevent substances reaching the developing spermatozoa (through drug transporters)
  • establish a basal and adluminal (apical) compartment (specialized microenvironment)
  • provide an immunological privilege status of the testis (anti-sperm antibodies are not developed)
Rat Blood–Testis Barrier
A model depicting the migration of developing preleptotene spermatocytes across the blood–testis barrier (BTB}.[15]
Rat blood–testis barrier 02.jpg Rat blood–testis barrier 03.jpg
Movement of developing germ cells in the seminiferous epithelium of the adult testis.

Germ cells first have to break through the tight junctions at the BTB with minimal disruptions. After the opening of the BTB, the progression of germ cells relies solely on series of transient adherens junctions at the Sertoli–germ cell interface. Assembly and disassembly of junctional complexes at the basal and apical ectoplasmic specialization occurs continuously.

When germ cells move along Sertoli cells, the original JAM/CAR/nectin trans-homodimers between Sertoli cells are replaced by the Sertoli–germ cell junctions composed of CAR–CAR/CAR–JAM-C/nectin-2–nectin-3/necl-5(PVR)–necl-2/JAM-B–JAM-C complexes plausibly through competitive binding. It is noted that CAR–CAR adhesion has not been directly demonstrated for Sertoli cell–germ cell interaction, but remains an attractive possibility.
Text from original figure legend.

Hormones

These cells secrete several hormones, anti-Müllerian hormone (AMH), inhibin and activin.

Inhibin and activin provide positive and negative feedback respectively on follicle-stimulating hormone (FSH) secretion from the pituitary.

  • activins - dimers of beta-A and/or beta-B subunits encoded by the genes INHBA and INHBB
  • Follicle-stimulating hormone (FSH)-releasing protein (FRP) subunit is identical in structure to the beta-A subunit of inhibin.

Anti-Mullerian Hormone

Anti-Mullerian Hormone (Anti-Müllerian Hormone, AMH) is a glycoprotein that is produced by immature Sertoli cells.

Anti-Mullerian hormone (AMH) is a glycoprotein belonging to the transforming growth factor-beta (TGF-beta) family. During development, immature Sertoli cells produce AMH[16] to aid regression of Müllerian (paramesonephric) ducts in male sex developement.[17]

Follicle-stimulating hormone (FSH) promotes AMH transcription in the absence of androgen signaling, while testosterone has been shown to inhibit the transcriptional activation of AMH.


Links: Anti-Mullerian Hormone

Abnormalities

Histology

Testis Histology Links: Testis Development | Spermatozoa Development | Histology

Human (young): overview labeled | overview unlabeled | convoluted seminiferous tubules x10 | x40 | x40 | tunica albuginea x20
Human (adult): overview x2 | convoluted seminiferous tubules labeled | x10 | x20 | x40 | x40 | epididymis ductulus efferens | ductus epididymidis | epithelium | overview x4 | x10 | x20 | x40 | ductus deferens labeled overview | epithelium | overview x2 | x10 | x40
Human spermatozoa: x20 | x40 | x100
Human Stage 22: Testis - labeled overview | Testis - unlabeled overview | Testis - unlabeled detail | Testis - labeled detail | testis | Carnegie stage 22 | Movie - Urogenital stage 22
Rabbit: convoluted seminiferous tubules x20 | x100
Mouse: postnatal epididymis | 14 days postnatal | 33 days postnatal | 45 days postnatal | 2 months postnatal
Spermatozoa Development (expand to see terms)  

Spermatozoa Development

Note there are additional glossaries associated with genital, spermatozoa, oocyte and renal.

Spermatozoon
  • acroplaxome - structure forms the acrosome plate with intermediate filament bundles of the marginal ring at the leading edge of the acrosome. The sub-acrosomal layer located in the developing spermatozoa head perinuclear region, located between the inner acrosomal membrane and the nuclear envelope. The other part of the perinuclear region is the post-acrosomal sheath (PAS) at the post-acrosomal region.
  • acrosome - Cap-shaped cellular structure formed from the golgi apparatus and contains enzymes to dissolve the oocyte (egg) zona pellucida for fertilisation.
  • acrosome compaction - Acrosome reshaping process in final stages of spermatogenesis (spermatid to spermatozoa).
  • acrosome reaction - Chemical change within the spermatozoa following binding to the zona pellucida, only acrosome reacted spermatozoa have an ability to fuse with oocytes.
  • annulus - Cytoskeletal (septin) structure located between the midpiece and principal piece regions of the tail, thought to form a diffusion barrier between these two domains. PMID 20042538
  • asthenozoospermia - (asthenospermia) Term for reduced sperm motility and can be the cause of male infertility.
  • axoneme - (axonema) The basic structure in cilia and eukaryotic flagella and in the spermatozoa tail, consisting of parallel microtubules in a characteristic "9 + 2" pattern. This pattern describes 9 outer microtubule doublets (pairs) surrounding 2 central singlet microtubules, in humans 50 μm long. The motor protein dynenin move the outer microtubules with respect to the central pair, bending the cilia and generating motility. Note that prokaryotic bacteria have a similar process (flagellum) that uses an entirely different mechanism for motility.
  • capacitation - term describing the process by which spermaozoa become capable of fertilizing an oocyte, requires membrane changes, removal of surface glycoproteins and increased motility.
  • caput - proximal head of the epididymis, epithelium with stereocilia, involved in absorbing fluid to concentrate spermatozoa. Underlying smooth muscle aids movement. Epididymis three main parts : caput (head), corpus (body), cauda (tail).
  • CatSper - cationic (Ca2+) channel of spermatozoa, progesterone activated involved in hyperactivation, acrosome reaction, and possibly chemotaxis.
  • cauda - distal tail of the epididymis, region with a thin epithelium and the greatest quantity of smooth muscle. Epididymis three main parts : caput (head), corpus (body), cauda (tail).
  • centriole - a microtubule organising centre. First required for axoneme formation (distal centriole) that is lost and a second for pronuclei formation (proximal) following fertilisation. Rodents loose both and only have maternal centrioles.
  • connecting piece - linkage between the spermatozoa head and the midpiece of the tail. PMID 22767409
  • corpus - elongated body of the epididymis, This has an intermediate thickness of epithelium and thicker smooth muscle layer than caput. Epididymis three main parts : caput (head), corpus (body), cauda (tail).
  • cytoplasmic bridges - Transient cytoplasm connections between spermatids arising from one spermatogonium due to incomplete cytokinesis.
  • diploid - (Greek, di = double + ploion = vessel) Having two sets of chromosomes, the normal state for all cells other than the gametes.
  • end piece - Last portion of the spermatozoa tail region.
  • epididymis - testis tubular structure connecting the efferent ducts to the ductus deferent and functions for the storage and maturation of spermatozoa. Epididymis three main parts : caput (head), corpus (body), cauda (tail). PMID27307387
  • fibrous sheath - cytoskeletal structure surrounding the axoneme and outer dense fibers, defining the extent of the principal piece region.
  • haploid - (Greek, haploos = single) Having a single set of chromosomes as in mature germ/sex cells (oocyte, spermatozoa) following reductive cell division by meiosis. Normally cells are diploid, containing 2 sets of chromosomes.
  • interstitial cell - (Leydig cell) Male gonad (testis) cell which secrete the androgen testosterone, beginning in the fetus.
  • interstitium - testis developmental region (space between testis cords) that generates Leydig cells and other less well characterized cell types.
  • Johnsen score - a clinical score (1-10) for assessing spermatogenesis in a human testicular biopsy. Named after the author of the original article. PMID 5527187
  • Leydig cell - (interstitial cell) Male gonad (testis) cell that secrete the androgen testosterone, beginning in the fetus. Fetal Leydig cells develop from coelomic epithelium and undifferentiated perivascular cells in the gonad–mesonephros border region. Adult Leydig cells appear after birth from stem/progenitor cells among peritubular and peri-vascular cells. Leydig cells were first histologically identified in 1850 by Franz von Leydig (1821 - 1908) a German scientist.
  • meiosis - The cell division that occurs only in production of germ cells where there is a reduction in the number of chromosomes (diploid to haploid) which is the basis of sexual reproduction. All other non-germ cells in the body divide by mitosis.
  • midpiece - (middle piece) spermatozoa tail initial segment of axoneme surrounded outer dense fibres then by mitochondria. Next in the tail is the principal piece then finally the end piece.
  • mitosis - The normal division of all cells, except germ cells, where chromosome number is maintained (diploid). In germ cell division (oocyte, spermatozoa) meiosis is a modified form of this division resulting in reduction in genetic content (haploid). Mitosis, division of the nucleus, is followed by cytokinesis the division of the cell cytoplasm and the cytoplasmic contents. cytokinesis overlaps with telophase.
  • outer dense fibres - (ODF, outer dense fibers) cytoskeletal structures that surround the axoneme in the middle piece and principal piece of the spermatozoa tail.
  • primary spermatocyte - arranged in the seminiferous tubule wall deep (luminal) to the spermatogonia. These large cells enter the prophase of the first meiotic division. (More? meiosis)
  • principal piece - Spermatozoa tail segment containing the plasma membrane calcium channels (CatSper1 and CatSper2) required for hyperactivation of motility. Region is partially separated from the midpiece by a barrier called the annulus.
  • sertoli cells - (sustentacular cell) These cells are the spermatozoa supporting cells, nutritional and mechanical, as well as forming a blood-testis barrier. The cell cytoplasm spans all layers of the seminiferous tubule. The cells are named after Enrico Sertoli (1842 - 1910), and italian physiologist and histologist.
  • sperm annulus - (Jensen's ring; Latin, annulus = ring) A region of the mammalian sperm flagellum connecting the midpiece and the principal piece. The annulus is a septin-based structure formed from SEPT1, 4, 6, 7 and 12. Septins are polymerizing GTPases that can act as a scaffold forming hetero-oligomeric filaments required for cytokinesis and other cell cycle roles.
  • spermatogenesis - (Greek, genesis = origin, creation, generation) The term used to describe the process of diploid spermatagonia division and differentiation to form haploid spermatazoa within the testis (male gonad). The process includes the following cellular changes: meiosis, reoorganization of DNA, reduction in DNA content, reorganization of cellular organelles, morphological changes (cell shape). The final process of change in cell shape is also called spermiogenesis.
  • spermatogenesis - (Greek, genesis = origin, creation, generation) The maturation process of the already haploid spermatazoa into the mature sperm shape and organization. This process involves reorganization of cellular organelles (endoplasmic reticulum, golgi apparatus, mitochondria), cytoskeletal changes (microtubule organization) and morphological changes (cell shape, acrosome and tail formation).
  • spermatogonia - The cells located in the seminiferous tubule adjacent to the basal membrane that either divide and separate to renew the stem cell population, or they divide and stay together as a pair (Apr spermatogonia) connected by an intercellular cytoplasmic bridge to differentiate and eventually form spermatazoa.
  • spermatozoa head - Following spermiogenesis, the first region of the spermatozoa containing the haploid nucleus and acrosome. In humans, it is a flattened structure (5 µm long by 3 µm wide) with the posterior part of nuclear membrane forming the basal plate region. The human spermatozoa is about 60 µm long, actively motile and divided into 3 main regions (head, neck and spermatozoa tail).
  • spermatozoa neck - Following spermiogenesis, the second region of the spermatozoa attached to basal plate, transverse oriented centriole, contains nine segmented columns of fibrous material, continue as outer dense fibres in tail. In humans, it forms a short structure (1 µm). The human spermatozoa is about 60 µm long, actively motile and divided into 3 main regions (head, neck and tail).
  • spermatozoa tail - Following spermiogenesis, the third region of the spermatozoa that has a head, neck and tail). The tail is also divided into 3 structural regions a middle piece, a principal piece and an end piece. In humans: the middle piece (5 µm long) is formed by axonema and dense fibres surrounded by mitochondria; the principal piece (45 µm long) fibrous sheath interconnected by regularly spaced circumferential hoops; the final end piece (5 µm long) has an axonema surrounded by small amount of cytoplasm and plasma membrane.
  • spermatogonial stem cells - (SSCs) The spermatagonia cells located beside the seminiferous tubule basal membrane that either divide and separate to renew the stem cell population, or they divide and stay together as a pair (|Apr spermatogonia) connected by an intercellular cytoplasmic bridge to differentiate and eventually form spermatazoa.
  • spermatozoon - singular form of of spermatozoa.
  • sperm protein 56 - A component of the spermatozoa acrosomal matrix released to the sperm surface during capacitation.
  • teratospermia - Clinical term for a spermatozoa with abnormal morphology (small, large, defects in the head, tail, and/or mid-piece) present in the semen or ejaculate.
  • testis cords - developmental structure that give rise to the adult seminiferous tubules, the other developmental region is the interstitium.
  • vasectomy - Clinical term for ligation of the scrotal portion of the ductus deferens.

See also: Spermatozoa Terms collapse table

Other Terms Lists  
Terms Lists: ART | Birth | Bone | Cardiovascular | Cell Division | Endocrine | Gastrointestinal | Genital | Genetic | Head | Hearing | Heart | Immune | Integumentary | Neonatal | Neural | Oocyte | Palate | Placenta | Radiation | Renal | Respiratory | Spermatozoa | Statistics | Tooth | Ultrasound | Vision | Historic | Drugs | Glossary
Genital Links: genital | Lecture - Medicine | Lecture - Science | Lecture Movie | Medicine - Practical | primordial germ cell | meiosis | endocrine gonad‎ | Genital Movies | genital abnormalities | Assisted Reproductive Technology | puberty | Category:Genital
Female | X | X inactivation | ovary | corpus luteum | oocyte | uterus | vagina | reproductive cycles | menstrual cycle | Category:Female
Male | Y | SRY | testis | spermatozoa | ductus deferens | penis | prostate | Category:Male
Historic Embryology - Genital 
General: 1901 Urinogenital Tract | 1902 The Uro-Genital System | 1904 Ovary and Testis | 1912 Urinogenital Organ Development | 1914 External Genitalia | 1921 Urogenital Development | 1921 External Genital | 1942 Sex Cords | 1953 Germ Cells | Historic Embryology Papers | Historic Disclaimer
Female: 1904 Ovary and Testis | 1904 Hymen | 1912 Urinogenital Organ Development | 1914 External Genitalia | 1914 Female | 1921 External Genital | 1927 Female Foetus 15 cm | 1927 Vagina | 1932 Postnatal Ovary
Male: 1887-88 Testis | 1904 Ovary and Testis | 1904 Leydig Cells | 1906 Testis vascular | 1909 Prostate | 1912 Prostate | 1914 External Genitalia | 1915 Cowper’s and Bartholin’s Glands | 1920 Wolffian tubules | 1935 Prepuce | 1935 Wolffian Duct | 1942 Sex Cords | 1943 Testes Descent | Historic Embryology Papers | Historic Disclaimer
Links: Spermatozoa Histology | Histology Stain H&E | Histology Stains

Molecular

Sry

  • Y chromosome gene for a transcription factor
  • member of the high mobility group (HMG)-box family of DNA binding proteins
  • human - 204 amino acid protein[18]
Links: OMIM - Sry

Sox9

  • autosomal transcription factor
  • Development of XY females - presence of only a single functional copy of the transcription factor encoding genes SOX9, SF1, or WT1 (Note- not all XY humans are sex-reversed if only a single copy of a normal SF1 or WT1 allele is present)
  • A nuclear export signal within the high mobility group domain regulates the nucleocytoplasmic translocation of SOX9 during sexual determination[19]

Other roles

  • Cartilage - essential for chondrocyte differentiation
  • Hearing - otic placode formation, maintenance of progenitors in the otic epithelium


Links: Sox9 | Cartilage Development | Inner Ear Development

Fog2

  • transcription factor, named Friend of Gata2
  • human - (8q23) 1,151 amino acid nuclear protein that contains 8 zinc finger motifs[20]
  • dosage critical for fetal testis development in mice[21]
Links: OMIM - Fog2

Gadd45g

Gadd45g and Sex Determination Model[6]

Growth Arrest- And Dna Damage-Inducible Gene (GADD45, GAMMA; GADD45G)

A Recent mouse study[6] has shown that Gadd45g protein has a role in primary sex differentiation. Knockout mice (Gadd45g(-/-) XY gonads) resulted in a a sex reversal.


Links: OMIM - Gadd45g

Gata4

  • transcription factor
  • dosage critical for fetal testis development in mice[21]

Eif2s3y

References

  1. Rijntjes E, Gomes MLM, Zupanič N, Swarts HJM, Keijer J & Teerds KJ. (2017). Transient Hypothyroidism: Dual Effect on Adult-Type Leydig Cell and Sertoli Cell Development. Front Physiol , 8, 323. PMID: 28588502 DOI.
  2. Sertoli E. (2018). The structure of seminiferous tubules and the development of [spermatids] in rats. Biol. Reprod. , 99, 482-503. PMID: 29961830 DOI.
  3. Baratelli GM, Lanzani A & Sacco RN. (2002). Biography of Enrico Sertoli. Urology , 60, 196-8. PMID: 12100962
  4. Wen Q, Wang Y, Tang J, Cheng CY & Liu YX. (2016). Sertoli Cell Wt1 Regulates Peritubular Myoid Cell and Fetal Leydig Cell Differentiation during Fetal Testis Development. PLoS ONE , 11, e0167920. PMID: 28036337 DOI.
  5. Rebourcet D, O'Shaughnessy PJ, Pitetti JL, Monteiro A, O'Hara L, Milne L, Tsai YT, Cruickshanks L, Riethmacher D, Guillou F, Mitchell RT, van't Hof R, Freeman TC, Nef S & Smith LB. (2014). Sertoli cells control peritubular myoid cell fate and support adult Leydig cell development in the prepubertal testis. Development , 141, 2139-49. PMID: 24803659 DOI.
  6. 6.0 6.1 6.2 Johnen H, González-Silva L, Carramolino L, Flores JM, Torres M & Salvador JM. (2013). Gadd45g is essential for primary sex determination, male fertility and testis development. PLoS ONE , 8, e58751. PMID: 23516551 DOI.
  7. Hiramatsu R, Harikae K, Tsunekawa N, Kurohmaru M, Matsuo I & Kanai Y. (2010). FGF signaling directs a center-to-pole expansion of tubulogenesis in mouse testis differentiation. Development , 137, 303-12. PMID: 20040496 DOI.
  8. Heyn R, Makabe S & Motta PM. (2001). Ultrastructural morphodynamics of human Sertoli cells during testicular differentiation. Ital J Anat Embryol , 106, 163-71. PMID: 11732573
  9. Karl J & Capel B. (1998). Sertoli cells of the mouse testis originate from the coelomic epithelium. Dev. Biol. , 203, 323-33. PMID: 9808783 DOI.
  10. Archambeault DR & Yao HH. (2010). Activin A, a product of fetal Leydig cells, is a unique paracrine regulator of Sertoli cell proliferation and fetal testis cord expansion. Proc. Natl. Acad. Sci. U.S.A. , 107, 10526-31. PMID: 20498064 DOI.
  11. Kopera IA, Bilinska B, Cheng CY & Mruk DD. (2010). Sertoli-germ cell junctions in the testis: a review of recent data. Philos. Trans. R. Soc. Lond., B, Biol. Sci. , 365, 1593-605. PMID: 20403872 DOI.
  12. Griswold MD. (1995). Interactions between germ cells and Sertoli cells in the testis. Biol. Reprod. , 52, 211-6. PMID: 7711190
  13. Cheng CY & Mruk DD. (2012). The blood-testis barrier and its implications for male contraception. Pharmacol. Rev. , 64, 16-64. PMID: 22039149 DOI.
  14. Su L, Mruk DD & Cheng CY. (2011). Drug transporters, the blood-testis barrier, and spermatogenesis. J. Endocrinol. , 208, 207-23. PMID: 21134990 DOI.
  15. Wang CQ & Cheng CY. (2007). A seamless trespass: germ cell migration across the seminiferous epithelium during spermatogenesis. J. Cell Biol. , 178, 549-56. PMID: 17698604 DOI.
  16. Josso N. (1992). Anti-müllerian hormone and Sertoli cell function. Horm. Res. , 38 Suppl 2, 72-6. PMID: 1292986 DOI.
  17. Rehman ZU, Worku T, Davis JS, Talpur HS, Bhattarai D, Kadariya I, Hua G, Cao J, Dad R, Farmanullah T, Hussain L & Yang. (2017). Role and mechanism of AMH in the regulation of Sertoli cells in mice. J. Steroid Biochem. Mol. Biol. , 174, 133-140. PMID: 28851672 DOI.
  18. Su H & Lau YF. (1993). Identification of the transcriptional unit, structural organization, and promoter sequence of the human sex-determining region Y (SRY) gene, using a reverse genetic approach. Am. J. Hum. Genet. , 52, 24-38. PMID: 8434602
  19. Gasca S, Canizares J, De Santa Barbara P, Mejean C, Poulat F, Berta P & Boizet-Bonhoure B. (2002). A nuclear export signal within the high mobility group domain regulates the nucleocytoplasmic translocation of SOX9 during sexual determination. Proc. Natl. Acad. Sci. U.S.A. , 99, 11199-204. PMID: 12169669 DOI.
  20. Holmes M, Turner J, Fox A, Chisholm O, Crossley M & Chong B. (1999). hFOG-2, a novel zinc finger protein, binds the co-repressor mCtBP2 and modulates GATA-mediated activation. J. Biol. Chem. , 274, 23491-8. PMID: 10438528
  21. 21.0 21.1 Bouma GJ, Washburn LL, Albrecht KH & Eicher EM. (2007). Correct dosage of Fog2 and Gata4 transcription factors is critical for fetal testis development in mice. Proc. Natl. Acad. Sci. U.S.A. , 104, 14994-9. PMID: 17848526 DOI.


Reviews

Rotgers E, Jørgensen A & Yao HH. (2018). At the crossroads of fate - somatic cell lineage specification in the fetal gonad. Endocr. Rev. , , . PMID: 29771299 DOI.

Barton LJ, LeBlanc MG & Lehmann R. (2016). Finding their way: themes in germ cell migration. Curr. Opin. Cell Biol. , 42, 128-137. PMID: 27484857 DOI.

De Felici M. (2016). The Formation and Migration of Primordial Germ Cells in Mouse and Man. Results Probl Cell Differ , 58, 23-46. PMID: 27300174 DOI.

Virtanen HE & Toppari J. (2014). Embryology and physiology of testicular development and descent. Pediatr Endocrinol Rev , 11 Suppl 2, 206-13. PMID: 24683945

Svingen T & Koopman P. (2013). Building the mammalian testis: origins, differentiation, and assembly of the component cell populations. Genes Dev. , 27, 2409-26. PMID: 24240231 DOI.

Hinton BT, Galdamez MM, Sutherland A, Bomgardner D, Xu B, Abdel-Fattah R & Yang L. (2011). How do you get six meters of epididymis inside a human scrotum?. J. Androl. , 32, 558-64. PMID: 21441421 DOI.

Griswold SL & Behringer RR. (2009). Fetal Leydig cell origin and development. Sex Dev , 3, 1-15. PMID: 19339813 DOI.

Cheng CY & Mruk DD. (2012). The blood-testis barrier and its implications for male contraception. Pharmacol. Rev. , 64, 16-64. PMID: 22039149 DOI.

Articles

Kaftanovskaya EM, Feng S, Huang Z, Tan Y, Barbara AM, Kaur S, Truong A, Gorlov IP & Agoulnik AI. (2011). Suppression of insulin-like3 receptor reveals the role of β-catenin and Notch signaling in gubernaculum development. Mol. Endocrinol. , 25, 170-83. PMID: 21147849 DOI.

Stukenborg JB, Colón E & Söder O. (2010). Ontogenesis of testis development and function in humans. Sex Dev , 4, 199-212. PMID: 20664245 DOI.

Hutson JM, Balic A, Nation T & Southwell B. (2010). Cryptorchidism. Semin. Pediatr. Surg. , 19, 215-24. PMID: 20610195 DOI.

Adham IM & Agoulnik AI. (2004). Insulin-like 3 signalling in testicular descent. Int. J. Androl. , 27, 257-65. PMID: 15379965 DOI.

Heyns CF. (1987). The gubernaculum during testicular descent in the human fetus. J. Anat. , 153, 93-112. PMID: 2892824

Books

De Groot LJ, Chrousos G, Dungan K, Feingold KR, Grossman A, Hershman JM, Koch C, Korbonits M, McLachlan R, New M, Purnell J, Rebar R, Singer F, Vinik A & Hutson JM. (2000). Cryptorchidism and Hypospadias. , , . PMID: 25905331


Skinner M. and Griswold M. Sertoli Cell Biology (2004) Academic Press

Search PubMed

Search Pubmed: Testis Development | Epididymis Development | Sry | Sox9 |

Additional Images

Historic Images

Historic Embryology: 1912 Urinogenital Organ Development | 1921 Urogenital Development | 1921 External Genital Development | Historic Disclaimer
Historic Disclaimer - information about historic embryology pages 
Mark Hill.jpg
Pages where the terms "Historic" (textbooks, papers, people, recommendations) appear on this site, and sections within pages where this disclaimer appears, indicate that the content and scientific understanding are specific to the time of publication. This means that while some scientific descriptions are still accurate, the terminology and interpretation of the developmental mechanisms reflect the understanding at the time of original publication and those of the preceding periods, these terms, interpretations and recommendations may not reflect our current scientific understanding.     (More? Embryology History | Historic Embryology Papers)

External Links

External Links Notice - The dynamic nature of the internet may mean that some of these listed links may no longer function. If the link no longer works search the web with the link text or name. Links to any external commercial sites are provided for information purposes only and should never be considered an endorsement. UNSW Embryology is provided as an educational resource with no clinical information or commercial affiliation.



Glossary Links

Glossary: A | B | C | D | E | F | G | H | I | J | K | L | M | N | O | P | Q | R | S | T | U | V | W | X | Y | Z | Numbers | Symbols | Term Link




Cite this page: Hill, M.A. (2024, April 16) Embryology Sertoli cell. Retrieved from https://embryology.med.unsw.edu.au/embryology/index.php/Sertoli_cell

What Links Here?
© Dr Mark Hill 2024, UNSW Embryology ISBN: 978 0 7334 2609 4 - UNSW CRICOS Provider Code No. 00098G