Role of connexin-based gap junction channels in testis

https://doi.org/10.1016/j.tem.2005.07.001Get rights and content

Spermatogenesis is a highly controlled process that allows proliferation and differentiation of male germ cells. This is under classical endocrine and paracrine controls. There is also evidence that gap junctions between Leydig cells, between Sertoli cells and between Sertoli and germ cells participate in the local regulation of spermatogenesis. Recent studies reveal that connexin 43 (Cx43), the predominant gap junction protein in the testis, is essential for the initiation and maintenance of spermatogenesis. In this review, we focus on the identification, distribution and control of connexins in the mammalian testis. The implication of connexin-based gap junctions in testicular physiology and in pathological disorders of spermatogenesis (spermatogenic arrest and testis cancer) is also discussed.

Section snippets

Gap junction channels and their constitutive proteins: connexins

Gap junctions are intercellular, plasma-membrane channels that link the cytoplasm of neighboring cells in numerous tissues except erythrocytes, platelets, fully differentiated skeletal muscle cells and sperm. They allow the direct exchange of molecules with a relative molecular mass up to 1 kDa, such as metabolic precursors, nutrients and second messengers including inositol (1,4,5)-triphosphate [Ins(1,4,5)P3], cAMP and ions [3]. This process of exchange of molecular signals, also termed

Gap junctions and Cxs in the testis

Gap junctions were identified first by electron microscopy in the testis of rodents before and during the initiation of spermatogenesis [10]. In the mature rat testis, transcripts for 11 Cxs have been identified (Cx26, Cx30.2, Cx31, Cx31.1, Cx32, Cx33, Cx37, Cx40, Cx43, Cx46 and Cx50), 10 of these are present in polysomes and are, presumably, translated [11].

GJIC occurs between Leydig cells, which express only Cx43 12, 13, 14. In seminiferous tubules, gap junctions are located between Sertoli

Physiological relevance of gap junctions and Cxs in the testis

The possibility that Cx43 plays a primordial role in gonadal development and the control of spermatogenesis results from observations in both fertile and mutant, sterile animals, and in mice in which the genes encoding Cxs are deleted. Cx43 forms intercellular contacts between Sertoli cells and germ cells in proliferation [20] and Cx43-based gap junctions appear to be controlled in a stage-dependent manner, which indicates a close relationship between the association of specific germ cells and

Control of Cx expression in the testis

Despite the well-documented presence and indispensability of Cx43, there is limited information regarding how GJIC and Cxs are regulated in the testis. A schematic illustration of Cx gap junction control in the testis is presented in Figure 2. There is evidence that the expression of Cx43 depends on the stage of the spermatogenic cycle in rat 15, 16, 19, 25, 47, 48 and human testes 17, 18. In mutant mice with impaired spermatogenesis, a close relationship between the presence of specific germ

Gap junctions and Cx43 expression in testicular diseases

Mutations in genes that encode Cxs result in human genetic disorders [7], and impaired GJIC and a lack of transcription of Cx genes correlate with neoplastic transformation in several human tissues 63, 64. To date, there is limited information on gap junctions in pathological testes. Freeze-fracture studies show no variation in the number of gap-junctional particles in the seminiferous tubules of azoospermic and oligospermic patients [65]. However, there is no information on the origin and

Concluding remarks and future perspectives

In addition to the classical functions of Sertoli cells, the data presented in this review highlight a new type of Sertoli–germ cell communication that appears to be essential for spermatogenesis. If the involvement of Sertoli–germ cell communication is associated mainly with the presence of Cx43, the function of native Cx33, which exhibits a dominant-negative effect on GJIC, remains to be defined. The lack of Cx33 ortholog in the human genome [7] does not, however, mean that another member of

Acknowledgements

This work was supported, in part, by grants from the Institut National de la Santé et de la Recherche Médicale (INSERM) and by Pfizer. The authors thank A. Malassine and J-M. Gasc for critically reading the manuscript.

References (70)

  • T.W. White et al.

    Gap junctions: Fates worse than death?

    Curr. Biol.

    (2000)
  • A. Cheng

    Gap junctional communication is required to maintain mouse cortical neural progenitor cells in a proliferative state

    Dev. Biol.

    (2004)
  • C. Stout

    Connexins: functions without junctions

    Curr. Opin. Cell Biol.

    (2004)
  • F. Grassi

    Putative second messengers affect cell coupling in the seminiferous tubules

    Cell Biol. Int. Rep.

    (1986)
  • J.M. Luk

    Identification of novel genes expressed during spermatogenesis in stage-synchronized rat testes by differential display

    Biochem. Biophys. Res. Commun.

    (2003)
  • C. Fiorini

    Sertoli cell junctional proteins as early targets for different classes of reproductive toxicants

    Reprod. Toxicol.

    (2004)
  • M. Mesnil

    Connexins and cancer

    Biol. Cell.

    (2002)
  • E. Bigliardi et al.

    Gap junctions between Sertoli cells in the infertile human testis

    Fertil. Steril.

    (1977)
  • J.C. Herve

    Diversity in protein-protein interactions of connexins: emerging roles

    Biochim. Biophys. Acta

    (2004)
  • P.T. Saunders

    Germ cell-somatic cell interactions during spermatogenesis

    Reprod. Suppl.

    (2003)
  • D.D. Mruk et al.

    Sertoli-Sertoli and Sertoli-germ cell interactions and their significance in germ cell movement in the seminiferous epithelium during spermatogenesis

    Endocr. Rev.

    (2004)
  • R. Bruzzone

    The cellular Internet: on-line with connexins

    Bioessays

    (1996)
  • P. Phelan et al.

    Innexins get into the gap

    Bioessays

    (2001)
  • G. Sohl et al.

    An update on connexin genes and their nomenclature in mouse and man

    Cell Commun. Adhes.

    (2003)
  • T.W. White

    Functional analysis of selective interactions among rodent connexins

    Mol. Biol. Cell

    (1995)
  • J.C. Saez

    Plasma membrane channels formed by connexins: their regulation and functions

    Physiol. Rev.

    (2003)
  • L.D. Russel

    Morphological and functional evidence for Sertoli-germ cell relationships

  • M.S. Risley

    Connexin gene expression in seminiferous tubules of the Sprague-Dawley rat

    Biol. Reprod.

    (2000)
  • W.A. Varanda et al.

    Intercellular communication between mouse Leydig cells

    Am. J. Physiol.

    (1994)
  • E.M. Perez-Armendariz

    Characterization of gap junctions between pairs of Leydig cells from mouse testis

    Am. J. Physiol.

    (1994)
  • M.S. Risley

    Cell-, age- and stage-dependent distribution of connexin43 gap junctions in testes

    J. Cell Sci.

    (1992)
  • C. Batias

    Modified expression of testicular gap-junction connexin 43 during normal spermatogenic cycle and in altered spermatogenesis

    Cell Tissue Res.

    (1999)
  • K. Steger

    Expression of connexin 43 in human testis

    Histochem. Cell Biol.

    (1999)
  • C. Batias

    Connexin43 gene expression and regulation in the rodent seminiferous epithelium

    J. Histochem. Cytochem.

    (2000)
  • X. Decrouy

    Functional characterization of Cx43 based gap junctions during spermatogenesis

    J. Cell. Physiol.

    (2004)
  • Cited by (101)

    • Effect of Bisphenol S on testicular tissue after low-dose lactation exposure

      2022, Environmental Pollution
      Citation Excerpt :

      It is well known that gap and tight junctions are the main components of the BTB. Therefore, we studied CX43, one of the well-known markers of gap junctions in the BTB (Li et al., 2010; Pointis & Segretain, 2005; Steger et al., 1999), and observed an increase in CX43 in adult males, with no difference in infants. This phenomenon can be explained by the fact that the BTB begins to develop from the first wave of spermatogenesis at the onset of puberty until the Sertoli cells’ proliferation is completed (Gerber et al., 2016).

    • Role of calcium oscillations in sperm physiology

      2021, BioSystems
      Citation Excerpt :

      Given that the synchronic activity is observed in large groups of cells, this work proposed that intercellular bridges could also be involved in the propagation of the [Ca2+]i oscillations between germ cells. During spermatogenesis cellular communication through gap junctions between Leydig cells, and between Sertoli – germ cells is fundamental for spermatogenesis regulation (Pointis and Segretain, 2005), and both gap junctions and intercellular bridges have been proposed to have an important role allowing synchronization, proliferation and differentiation of germ cells (Decrouy et al., 2004; Fawcett et al., 1959). Given that [Ca2+]i oscillations can regulate intracellular signaling mechanisms as kinases and phosphatases and also transcription factors (Smedler and Uhlén, 2014), our group proposed that during germ cell differentiation the [Ca2+]i oscillations transmitted through gap junctions and intracellular bridges could regulate both transduction and transcription elements maintaining groups of germ cells in the same functional and differentiation state into syncytium connected by intercellular bridges.

    View all citing articles on Scopus
    View full text