Gap junctions (GJs) provide for a unique system of intercellular communication (IC) allowing rapid transport of small molecules from cell to cell. GJs are formed by a large family of proteins named connexins (Cxs). Cx43 has been considered as the predominantly expressed Cx by hematopoietic-supporting stroma. To investigate the role of the Cx family in hemopoiesis, we analyzed the expression of 11 different Cx species in different stromal cell lines derived from murine bone marrow (BM) or fetal liver (FL). We found that up to 5 Cxs are expressed in FL stromal cells (Cx43, Cx45, Cx30.3, Cx31, and Cx31.1), whereas only Cx43, Cx45, and Cx31 were clearly detectable in BM stromal cells. In vivo, the Cx43-deficient 14.5- to 15-day FL cobblestone area–forming cells (CAFC)-week 1-4 and colony-forming unit contents were 26%-38% and 39%-47% lower than in their wild-type counterparts, respectively. The reintroduction of the Cx43 gene into Cx43-deficient FL stromal cells was able to restore their diminished IC to the level of the wild-type FL stromal cells. In addition, these Cx43-reintroduced stromal cells showed an increased support ability (3.7-fold) for CAFC-week 1 in normal mouse BM and 5-fold higher supportive ability for CAFC-week 4 in 5-fluorouracil-treated BM cells as compared with Cx43-deficient FL stromal cells. These findings suggest that stromal Cx43-mediated IC, although not responsible for all GJ-mediated IC of stromal cells, plays a role in the supportive ability for hemopoietic progenitors and stem cells.

Mature blood cells are derived from undifferentiated stem and progenitor cells in a highly complex series of maturational and divisional steps that occur in different tissues during embryonic development. The microenvironment seems to be an important factor influencing the proliferative activity and differentiation process of the stem and progenitor cells by local positive and negative signaling to the target cells.1 The following 3 mechanisms can be assumed to mediate the regulation of proliferation and differentiation of hematopoietic stem cells by stromal cells2: (a) by cytokine receptor-ligand interaction, (b) by interaction of adhesion molecules on hemopoietic cells with stromal cells or the extracellular matrix, or (c) by direct cell-to-cell communication between stromal cells or between stromal cells and hemopoietic cells.

Very little is known about the regulatory mechanisms of direct cell-to-cell communication in the hematopoietic microenvironment. Intercellular gap junctions (GJs) represent the most well-known intercellular communication (IC) system, and they are characterized by the existence of plaques of narrow channels between contacting cells. Each channel is formed by 2 hemichannels or hemiconnexons, and each one of them is contributed by 1 of the 2 adjacent cells. A hemiconnexon is an oligomeric assembly of 6 polypeptide subunits, or connexins (Cxs). Different tissue-specific Cxs have been characterized and cloned.3 These proteins are highly homologous and encoded by at least 4 gene subfamilies (α, β, γ, and δ)4,5that include 15 different Cxs in rodents. According to their molecular weight, they have been named Cx26, Cx30, Cx30.3, Cx31, Cx31.1, Cx32, Cx33, Cx36, Cx37, Cx40, Cx43, Cx45, Cx46, Cx50, and Cx57. These channels form the only known system for direct diffusional exchange of ions and small molecules (ie, molecular weight < 1000) between contacting cells.6 Nutrients and second messengers can be quickly transported in this way through cell communication networks.

There are few reports analyzing the GJs in hematopoietic tissues. By analysis of lucifer yellow (LY) dye transfer in a hemopoiesis-supporting bone marrow (BM) stromal cell line, Dorshkind et al7 showed the presence of Cx43-type GJ-IC that was inhibited by interleukin-1. Rosendaal et al8 reported on the basis of immunohistological studies that, in adult mouse BM, the Cx43 GJ epitopes are rare but up-regulated 80- to 100-fold in the marrow of the neonate or after forced stem cell division (by administering 5-fluorouracil [5-FU] or irradiation). This up-regulation occurred soon after the insult, before recognizable blood cells formed, and around the time at which primitive stem cells were triggered to go into cycle, suggesting the presence of a latent network of GJs in normal hemopoietic tissues. Our group9 has previously reported that the global blockade of all GJ-IC by amphotericin B reversibly inhibited the cobblestone area (CA) formation and hemopoiesis in stroma-containing cultures.

Strong evidence supports that more than 1 Cx gene is expressed in many specific tissues or cells. This fact suggests that different GJ proteins are allowing the permeability of different molecules. In osteoblasts10,11 and in fetal fibroblasts,12 it has been shown that different GJ proteins create channels with different conductance properties. This finding could suggest that their regulatory cell functions might also differ among them.

We conducted our efforts to study whether other Cxs, in addition to Cx43, are expressed by hemopoiesis-supporting stromal cells and whether stromal Cx43-dependent GJ-IC may influence hemopoiesis in vivo and in vitro in stroma-supported long-term culture.

Fetal liver and BM cells obtention

Heterozygous C57Bl/6-Gja-1tm1Kdr mice13 were housed under specified pathogen-free conditions and supplied with standard pellet food and water ad libitum. Mice were mated to produce Cx43 knockout (KO) embryos. Females were scored daily for the presence of the vaginal plug characteristic of the beginning of pregnancy. Day 0 of pregnancy was defined as the day of plug discovery (day 0 postcoitum) and the fetal specimens aged accordingly. Pregnant females were killed by cervical dislocation on day 14.5-15 of pregnancy, and the fetuses were removed through an anterior abdominal incision. After individual fetuses were isolated, fetal livers (FLs) and tails were carefully dissected to avoid contamination of surrounding tissues and were processed separately. Tails were used for Cx43 gene typing purposes. FL cells were suspended by pipetting the tissue several times in a medium consisting of calcium/magnesium-free phosphate-buffered saline (PBS; Gibco, Breda, The Netherlands), bovine serum albumin 0.5% (BSA; Sigma, St. Louis, MO), and sodium azide 0.01% (Merck, Darmstad, Germany; PBS/BSA/NaN3 buffer), which has been shown to maintain hematopoietic progenitors for 24-48 hours,14 until tail genotyping was finished (< 16 hours).

C57/Bl6, 10- to 15-week-old mice (from the Experimental Animal Facility, Erasmus University of Rotterdam, The Netherlands) were kept in the same conditions as mentioned above. In each experiment, 3-5 mice were intravenously injected with 150 mg/kg body weight 5-FU (Sigma) on day 0. Femoral BM was harvested on day +6 postinjection from normal and treated mice.

Stromal cell lines

We used 4 BM-derived stromal cell lines (FBMD-1, NBM11F4G, NBM11F10H, and NBM6D3D) and 4 FL-derived stromal cell lines (FL1E2C, FL7H7B, FLB6-1, which are all wild type (wt) for Cx43 gene—Cx43-wt cell lines—and FLB6-2 from Cx43-KO mice—Cx43 cell line). Except for the FBMD-1, FLB6-1, and FLB6-2 cell lines, the lines were cloned by limiting dilution and obtained from Dr T. Ghayur (BASF, Worcester, MA). Previously, we described that the FBMD-1, NBM11F4G, NBM11F10H, NBM6D3D, FL1E2C, and FL7H7B cell lines are able to support hemopoiesis in vitro and their differences in GJ-IC abilities, as assessed by LY dye transfer.9,15 The FLB6-1 and FLB6-2 stromal cell lines were developed by culturing cell explants from Cx43-wt and Cx43-KO day 15 FL from C57/Bl6-Gja1tm1Kdr mice, respectively. Genotyping for identification of homozygous mice was done by standard polymerase chain reaction (PCR) technique by using primers addressed to neomycin-phosphotransferase and Cx43 genes as previously described.16 Cell explants were cultured in T25 tissue-culture flasks (Costar, Corning, NY) at 33°C and 10% CO2. Culture medium contained IMDM, glutamax 5 mmol/L, 100 IU/mL penicillin, 0.1 mg/mL streptomycin (all from Gibco), 20% fetal calf serum (FCS; optimal batch for fibroblast-like cell growth; Hyclone Laboratories Inc., Logan, UT), 10 μmol/L hydrocortisone (hydrocortisone-21-hemisuccinate sodium salt; Sigma), and 100 μmol/L β-mercaptoethanol (Merck). Every 2 days, all medium was harvested and centrifuged, and a 1:1 mixture of conditioned medium and fresh medium was reinoculated into the T25 flasks. Clusters of adherent cells were rapidly seen after 2-4 days of culture. Adherent cells were trypsinized, and cells were submitted to at least 15 passages to be certain that they did not contain any hematological growth. These conditions strongly favor the outgrowth of fibroblastic cells. Once the cell lines were established, they were grown in a medium containing IMDM, 10% FCS, 5% horse serum, 100 IU/mL penicillin, 0.1 mg/mL streptomycin, and 10 μmol/L β-mercaptoethanol (stromal cell medium) at 33°C and 10% CO2. Cell lines were used for stromal support when fully confluent and between passages 15 and 21.

FLB6-1 showed spindle-shaped cells with thin, long cytoplasmic prolongations. These cells displayed contact inhibition, and their growth rate was similar to our BM reference stromal cell line FBMD-1. Phenotypic analysis of the FLB6-1 cell line showed positivity for Ly6A/E, CD34, and Ly6C but no membrane expression of CD3, B220, Mac-1, Gr-1, Thy-1, c-kit, or PECAM-1/CD31. The Cx43-KO cells (FLB6-2) present many short cytoplasmic prolongations, and they grew faster and at higher saturation density than the Cx43-wt stromal cells. Cx43-KO cells showed the same immunophenotype as Cx43-wt FL stromal cells.

Reintroduction of the Cx43 gene into Cx43-KO FL-derived stromal cells

We used the packaging cell line PA317/pBABECx43a (kindly provided by Dr Warn-Cramer and Dr Lau, University of Hawaii at Manoa, Honolulu, HI) because of its ability to reintroduce the Cx43 gene into mouse Cx43-deficient fibroblasts.12 The vector inserted in this cell line is the pBABECx43a, composed of the fragment G2 of the rat Cx43 gene as reported by Beyer et al,17 preceded by a 5′-LTR and a gag region and followed by a SV40 promoter that regulates the expression of the puromycin resistance gene. The cell line PA317/pBABECx43a was grown at 37°C, 5% CO2 in IMDM supplemented with 10% newborn calf serum, 100 IU/mL penicillin, 0.1 mg/mL streptomycin, and 7 μg/mL puromycin (Sigma) until 80% confluency. At that time, medium was changed for stromal cell medium with no puromycin. After 24 hours, supernatant was harvested and passed through a 0.45-μm mesh filter. Filtered retroviral supernatant was added to 10%-20% confluent passage 14 Cx43-KO FL stromal cells. Three successive daily incubations were performed in the same way until target cells reached 90% confluency. Thirty-six hours after starting the last cycle of incubation of target cells with the retroviral supernatant, medium was changed for stromal cell medium containing 7-10 μg/mL puromycin. Two cell lines were developed: FLB6-3 and FLB6-4 (Cx43+ cell lines). Transduced cells were passed for 7 additional passages to passage 21 with medium containing puromycin to maintain the transduced cells.

Calcein dye transfer among stromal cells

Calcein dye coupling of stromal cells was analyzed according to Ziambaras et al,18 with some modifications. Confluent stromal cells were trypsinized and incubated with 5 μmol/L calcein-am (Molecular Probes, Leiden, The Netherlands) for 30 minutes at 37°C. Cells were washed for 4 times in Ca++/Mg2+-containing Hanks balanced salted solution and passed through a 70-μm mesh filter. After that, 50 cells (in 0.5 mL stromal medium) were cocultured in each of 3 2-cm2 wells containing a pregrown homocellular confluent layer of stromal cells (around 40 000 cells per well). Wells were examined after 2 hours of coculture under an inverted fluorescent microscope. A positive event of GJ-IC was considered when a cluster of 2 or more neighboring cells was observed. A negative communicating event was considered when a single fluorescent cell was seen. The percentage of positive events and the number of fluorescent cells per cluster was recorded. Control incubation of stromal cells with 5 μmol/L calcein did not produce similar clusters, and it only increased background fluorescence in peripheral cells close to the edge of the well.

Immunofluorescence analysis for Cx proteins

Stromal cells were plated in culture chambers (Lab-Tek, Chamber Slide, Nunc, Naperville, IL) and grown overnight in stromal cell medium. After harvesting medium, slides were washed with PBS twice and fixed in ice-cold methanol for 30 minutes. After fixation, cells were permeabilized in 1% Triton-X-100, washed with PBS, and then incubated for 1 hour at room temperature in PBS/10% mouse serum containing the polyclonal rabbit antiserum directed against Cx4317 (1:70; Zymed, South San Francisco, CA), Cx4519 (1:150; Chemicon International, Temecula, CA), and Cx3120 (1:150; a small aliquot was kindly provided by Prof K. Willecke, University of Bonn, Germany). All of the antisera are able to recognize specific intracellular carboxy-terminal domains of the mentioned Cx proteins. After washing the slides, they were incubated with goat anti-rabbit FITC conjugate (Sigma) for 1 hour in the dark. After additional washing steps, the slides were mounted in Vectashield medium (Vector Laboratories, Burlingame, CA) and visualized by epifluorescence light microscopy.

Western blot assay for membrane-bound Cx43 expression

Flasks containing confluent cells were briefly rinsed with cold calcium/magnesium-free PBS and then lysed directly by addition of ice-cold alkaline buffer, containing 2 mmol/L NaHCO3, 20 mmol/L NaOH, 5 mmol/L EDTA and protease inhibitor Complete (Boehringer Mannheim, Mannheim, Germany) according to manufacturer's instructions. The lysate was sheared by serial passage through a 25-gauge needle and further homogenized by serial cycles of freezing and thawing. Protein concentration was determined by BCA protein assay (Pierce, Rockford, IL). The insoluble fragments enriched for membrane-bound Cx43 were collected after centrifugation for 60 minutes at 55 000 rpm at 4°C, resuspended in Laemmli buffer, and stored at −20°C until use.

Original protein (100 μg) was separated by SDS-PAGE (10% polyacrylamide) and transferred onto a nitrocellulose membrane (Protran BA83, Schleicher & Schuell, Dassel, Germany) by electroblotting. The membrane was incubated for 2 hours in 5% nonfat dry milk powder (Biorad, Veenendaal, The Netherlands) in PBS/0.1% Tween-20 (Sigma) for blocking nonspecific sites, rinsed in PBS, and incubated overnight with the polyclonal antibody anti-Cx43 (Zymed; 1:250) in PBS/10% FCS/0.1% Tween-20. The membrane was then washed 4 times with PBS/10% FCS/0.1% Tween-20 and incubated with a secondary antibody (antirabbit immunoglobulin horseradish peroxidase conjugate; 1:3000), and the bands were visualized with a chemoluminescence detection system according to manufacturer's instructions (ECL, Amersham-Pharmacia Biotech, Rainham, UK).

Reverse transcription (RT)-PCR

Monolayer confluent cell lines were lysed with Trizol (Life Technologies, Grand Island, NY) according to the manufacturer's instructions. RNA concentration was determined spectrophotometrically at 260 nm as previously described.21 RNAse inhibitor (120 IU) was added per 10 μg RNA, and RNAs were frozen at −80°C until use. In our experience, contaminating genomic DNA was still present in most samples. For that reason, we used a technique based on the use of Mn2+-DNAse to degrade contaminating genomic DNA,22 with some modifications. Briefly, a first 30-minute incubation of 10 μg of total RNA dissolved in 50 mmol/L Tris-HCl pH 8.3, 75 mmol/L KCl, 3 mmol/L MgCl2, 5 mmol/L MnCl2, 50 pg rabbit α-globin messenger RNA (mRNA; 90% purity; Gibco BRL) as an internal standard, and 5 IU RNAse-free DNAse I (Boehringer Mannheim) (total volume, 20 μL) was performed at 37°C. The reaction was stopped by heating at 70°C for 10 minutes, and the mix was split into 2 (−RT and +RT) tubes. A 10-μL solution containing oligo(dT)12-18 as a random primer (Pharmacia Biotech; final concentration: 100 μmol/L), dithiothreitol (Life Technologies; final concentration: 10 mmol/L), and each of the 4 dNTPs (Pharmacia Biotech; final concentration of each: 125 μmol/L) diluted in the same RT buffer were added. After 5 additional minutes at 72°C, 5 IU RT SuperScript Rnase H (Gibco BRL) was exclusively added to the +RT tubes. The reaction mixtures were incubated at 37°C during 90 minutes and at 92°C during 10 minutes for enzyme inactivation. Complementary DNAs (cDNAs) were used immediately or kept at −80°C until use.

cDNA aliquots corresponding to 0.25 μg of total RNA from the cell lines and 2.5 pg rabbit α-globin mRNA were amplified by using specific primers for 11 Cx genes23 and for the gene of hypoxanthine phosphoribosyltransferase (HPRT) (primers were 5′-GTAATGATCAGTCAACGGGGGAC-3′ and 3′-ACCAATTCCAACGTTCGAACGACC-5′; amplicon length: 179) as a housekeeping gene. Every PCR tube contained 10 mmol/L Tris-HCl, 50 mmol/L KCl, 1.5 mmol/L MgCl2, pH 8.3; 240 μmol/L dNTPs; 50 pmol of each primer of 5′-and 3′-Cx primers, 5′-rabbit α-globin and 3′-rabbit α-globin primers; and 1 unit of Taq DNA polymerase (SuperTaq, Sphaero-Q, Leiden, The Netherlands). The reactions were performed by using a touchdown protocol23 for 35 cycles. Control −RT tubes were always run in parallel. PCR products were incubated with the fluorescent dye Sybr-Green (Molecular Probes) and electrophoresed in a 2% agarose gel. Amplified DNA products were exposed to UV light, and green fluorescent-emitted light image was digitalized and recorded. Semiquantitative analyses were performed by determining the mean density ratio between the Cx and the rabbit α-globin bands in the PCR products of 3 2-fold serial dilutions of the cDNA.

Cobblestone area–forming cell assay

Cobblestone area–forming cell (CAFC) frequency analysis was performed as described previously.24 Stromal layers were prepared using the FBMD-1,15 Cx43(FLB6-2), and Cx43+ (FLB6-3 and FLB6-4) FL stromal cell lines. FBMD-1 cells were used between passages 16 and 20, FLB6-2 cells were used at passage 21, and the Cx43+ cell lines at passage +7 after the transduction (final passage 21). Flat-bottom 96-well plates (Falcon, Lincoln Park, NJ) were inoculated with 103 stromal cells per well from logarithmic phase cultures. Culture plastics destined for establishment of stromal feeders were incubated at 4°C overnight with 0.3% porcine gelatin (Sigma) in Milli-Q water to improve adherence of the stromal layer for up to 6 weeks. Except for the FBMD-1 cell line, the cell lines were irradiated to prevent overgrowth of the cells after reaching confluency. It has been demonstrated previously that BM stromal cell gap-junctional communication is resistant to irradiation in vitro.25 FL stromal cells were irradiated at 40 Gy delivered by a 137Cs source (Gammacell, Atomic Energy of Canada, Ottawa, Canada), 18-24 hours before inoculation. Suspensions of normal BM cells were overlaid on these stromal layers in 12 dilutions, 2-fold apart, consisting of 15 wells per dilution to allow limiting dilution analysis of the precursor cells forming hemopoietic clones under the stromal layer. Cultures were fed weekly by changing half of the medium and frequencies of CAFC determined at weekly intervals (CAFC-week 1 to CAFC-week 6). Wells were scored positive if at least one phase-dark hematopoietic clone (CA, containing 5 or more cells) was seen. The frequency of CAFC was then calculated by using Poisson statistics as described previously.2 6 

Colony-forming cell assay

FL cells (5 × 104 and 105) were plated for quantification of colony-forming unit granulocyte-macrophage (CFU-GM) and burst-forming unit erythroid (BFU-E) in a semisolid (0.88% methylcellulose; Methocel, Stade, Germany) culture medium (CellGro SCGM, Boehringer Ingelheim Bioproducts, Heidelberg, Germany). The erythroid cultures contained 30% FCS, 10% hemin (Sigma), 10 U/mL rh-erythropoietin (Jansen-Cilag, Tilburg, The Netherlands), and 50 ng/mL rm-SCF (kindly provided by Genetics Institute, Cambridge, MA). Colonies were scored after 10 days of culture at 37°C and 10% CO2.

Statistical analysis

Results are expressed as mean ± standard deviation except when otherwise stated. Baseline comparisons used Mann-Whitney U test for 2 independent samples, Kruskal-Wallis test for 3 or more independent samples, or Wilcoxon signed rank test for paired data. Statistical significance threshold was established atP < .05.

Cx gene expression

To characterize the Cx species expressed by stromal cells, we analyzed 4 BM-derived and 4 FL-derived cell lines able to support hemopoiesis in vitro. Of 11 Cx genes that we analyzed for mRNA transcription by RT-PCR, the expression of Cx26, Cx32, Cx37, Cx40, Cx46, and Cx50 was undetectable.

Figure 1 shows the expression of Cx30.3, Cx31, Cx31.1, Cx43, and Cx45 by RT-PCR. Cx45 was expressed by all of the cell lines. Cx43 was expressed by all of them except Cx43 (FLB6-2) cells as expected. Cx30.3 and Cx31 were expressed by all FL cell lines and faintly by NBM11F10H and NBM6D3D. Cx31 was expressed by all cell lines except the BM stromal cell line FBMD-1. By semiquantitative analysis of sets of 3 serial 2-fold dilutions of cDNA, we were not able to consistently observe any significant compensation in the expression of Cx45, Cx30.3, Cx31, or Cx31.1 in the Cx43 cell line with respect to all the Cx43-wt FL cell lines (data not shown).

Fig. 1.

Semiquantitative Cx mRNA expression in stromal cell lines.

The experiment shows the expression of Cx30.3 (182 bp, A), Cx31 (182 bp, B), Cx31.1 (357 bp, C), Cx43 (294 bp, D), and Cx45 (444 bp, E) in 8 different stromal cell lines. Rabbit α-globin was used as an internal standard (257 bp). HPRT was used as a housekeeping control gene (179 bp, F). Analyses were performed at least twice. Lane 1, FLB6-1; lane 2, FLB6-2; lane 3, FBMD-1; lane 4, NBM11F4G; lane 5, FL1E2C; lane 6, FL7H7B; lane 7, NBM11F10H; and lane 8, NBM6D3D.

Fig. 1.

Semiquantitative Cx mRNA expression in stromal cell lines.

The experiment shows the expression of Cx30.3 (182 bp, A), Cx31 (182 bp, B), Cx31.1 (357 bp, C), Cx43 (294 bp, D), and Cx45 (444 bp, E) in 8 different stromal cell lines. Rabbit α-globin was used as an internal standard (257 bp). HPRT was used as a housekeeping control gene (179 bp, F). Analyses were performed at least twice. Lane 1, FLB6-1; lane 2, FLB6-2; lane 3, FBMD-1; lane 4, NBM11F4G; lane 5, FL1E2C; lane 6, FL7H7B; lane 7, NBM11F10H; and lane 8, NBM6D3D.

Close modal

In addition, by using appropriate antibodies with known specificity, we studied the protein expression of 3 different Cx gene products (Cx43, Cx45, and Cx31) that were transcribed in both BM and FL cell lines. As observed by immunofluorescence, the expression of Cx43 was present in both BM and FL cell lines, but the expression pattern was different among cell lines (Table 1). In general, the plasma membrane showed most of the Cx43 expression (Figure2). This expression was frequently at the most distal points of the cell extensions, probably in regions of cell–cell contact. Cx expression on the membrane was either in a punctate way or more frequently as thick fluorescent spots, which could represent gap junctional plaques of varying size. In addition, FLB6-1 cells expressed a large amount of protein in the perinuclear/nuclear area with frequently brighter locations, suggesting large Cx43 protein pools destined to be readily expressed in the outer membrane. FL1E2C and FL7H7B cell lines also expressed Cx43 protein in either cell membrane or cytoplasm but at lower intensity as compared with FLB6-1, FBMD-1, and NBM6D3D (which had the highest level of expression). As expected, the fluorescence in FLB6-2 (Cx43-KO) cells for Cx43 staining was similarly faint as that found in the isotypic control.

Fig. 2.

Cx expression assessed by immunofluorescence.

Cx43, Cx45, and Cx31 expression as assessed by immunofluorescence in Cx43-wt (FLB6-1) and Cx43 (FLB6-2, from Cx43-KO mice) FL cells (OM: × 630). Note that fluorescence (arrows) is distributed in a punctate way or in thick fluorescent spots at the plasma membrane, frequently at the most distal points. Variable expression is also observed at the perinuclear area. Note that Cx43 and Cx31 expression form thicker fluorescent spots than Cx45 meanwhile. GAR-FITC, goat antirabbit FITC conjugate. See “Results” for additional description.

Fig. 2.

Cx expression assessed by immunofluorescence.

Cx43, Cx45, and Cx31 expression as assessed by immunofluorescence in Cx43-wt (FLB6-1) and Cx43 (FLB6-2, from Cx43-KO mice) FL cells (OM: × 630). Note that fluorescence (arrows) is distributed in a punctate way or in thick fluorescent spots at the plasma membrane, frequently at the most distal points. Variable expression is also observed at the perinuclear area. Note that Cx43 and Cx31 expression form thicker fluorescent spots than Cx45 meanwhile. GAR-FITC, goat antirabbit FITC conjugate. See “Results” for additional description.

Close modal

Immunolocalization of Cx43 in transduced cells demonstrated punctate staining of Cx43 in long cellular extensions and in regions of the plasma membrane (Figure 3A). However, as compared with Cx43-wt FL stromal cells, Cx43-reintroduced FL cells showed a fainter expression in their membranes and a more diffuse cytoplasmic fluorescent pattern. We quantitated the total Cx43 protein by immunoblot analysis of Cx43+ cell lines, revealing a reactive band with a molecular weight between 43 and 47 kD, as previously described.27 Cx43+ cell lines (FLB6-3 and FLB6-4) showed a level of expression of 23.7% and 9.1%, respectively, as compared with Cx43 highly expressing wt FL cells (FLB6-1, Figure 3B) that was in line with the immunocytologic analysis.

Fig. 3.

CX43 expression in FL stromal cells.

(A) Cx43 expression in the cell lines Cx43+ (FLB6-3 and FLB6-4, from Cx43-KO mice in which the Cx43 gene has been reintroduced). Note that cells show a great variance in their protein expression. Note that the fluorescence (similarly as in Cx43-wt cells, see Figure 2) is distributed along the membrane and the perinuclear area. (B) Western blot analysis of FL stromal cells. FLB6-1 (Cx43-wt) was chosen as Cx43-highly expressing cell line for comparison with Cx43 cells (FLB6-2) showing some background and Cx43+ cell lines (FLB6-3 and FLB6-4, representing 23.7% and 9.1% of the band density of FLB6-1, after background subtraction—FLB6-2).

Fig. 3.

CX43 expression in FL stromal cells.

(A) Cx43 expression in the cell lines Cx43+ (FLB6-3 and FLB6-4, from Cx43-KO mice in which the Cx43 gene has been reintroduced). Note that cells show a great variance in their protein expression. Note that the fluorescence (similarly as in Cx43-wt cells, see Figure 2) is distributed along the membrane and the perinuclear area. (B) Western blot analysis of FL stromal cells. FLB6-1 (Cx43-wt) was chosen as Cx43-highly expressing cell line for comparison with Cx43 cells (FLB6-2) showing some background and Cx43+ cell lines (FLB6-3 and FLB6-4, representing 23.7% and 9.1% of the band density of FLB6-1, after background subtraction—FLB6-2).

Close modal

Membrane Cx45 was more homogeneously found in the cell membranes of almost all cell lines studied and usually occurred in a very punctate way with occasional brighter local fluorescent spots and medium perinuclear/nuclear intensity (Figure 2). Cx31 expression was analyzed only in FL cells, and it was more similar to that of Cx43 than to Cx45. Fluorescent patches were more frequent in membrane contact areas as compared with Cx45. Cx31 expression pattern, in either Cx43-wt or Cx43 FL stromal cells, showed some occasional enlarged fluorescent areas and weaker cytoplasmic/perinuclear positivity than observed for Cx45 (Figure 2).

Calcein-dye transfer in BM and FL cell lines

All wt cell lines analyzed were able to transfer calcein at a similar level (Figure 4) with NBM11F10H showing some lower IC (ie, 63% of IC as compared with FBMD-1; Figure4). Among the FL cell lines, Cx43 stromal cells (FLB6-2) showed 65.8% of IC events (60.8% IC events with < 5 cells, 33.1% IC events with 5-10 cells, and 40.0% IC events with > 10 cells) as compared with the mean of their Cx43-wt FL counterparts (FL1E2C, FL7H7B, and FLB6-1). The reintroduction of the Cx43 gene (FLB6-3 and FLB6-4 cell lines) almost fully restored the level of IC at the different distance levels analyzed (Figure 4). The level of IC restoration was higher in the cell line FLB6-3 that expressed higher levels of Cx43 as indicated before.

Fig. 4.

Homocellular calcein dye transfer in 10 different stromal cell lines from BM and FL.

Two independent experiments with duplicate or triplicate wells per cell line were carried out. *P < .05, between NBM11F10H and the other BM cell lines and between the total IC level of Cx43 (FLB6-2) and the other FL cell lines.

Fig. 4.

Homocellular calcein dye transfer in 10 different stromal cell lines from BM and FL.

Two independent experiments with duplicate or triplicate wells per cell line were carried out. *P < .05, between NBM11F10H and the other BM cell lines and between the total IC level of Cx43 (FLB6-2) and the other FL cell lines.

Close modal

Content of CAFC and CFU in FL from Cx43-wt and Cx43-KO FL

To investigate whether the deletion of Cx43 would affect the hemopoietic development in the fetuses, we analyzed the CAFC (as assayed on the FBMD-1 cell line), CFU-GM, and BFU-E FL contents. In Cx43-KO mice, the content of all CAFC-week type subsets was decreased as compared with wt age-matched fetuses, with significant differences found for the CAFC-week 1-4 (62%-74% of their content in wt FLs; Figure 5A). The contents of total CFU-C and BFU-E per Cx43-KO FL were 61.3% and 53.0% of their respective content in wt FLs (Figure 5B).

Fig. 5.

Analysis of hemopoietic progenitor FL content from 14.5- to 15-day FLs from Cx43-wt (empty squares and bars) and Cx43-KO (filled squares and bars).

The results represent 3 independent experiments in which Cx43-wt (n = 11 fetuses; median: 7) and Cx43-KO (n = 9 fetuses; median: 3) FL cells had been obtained and compared within every experiment. (A) Weekly CAFC content. Actual mean numbers for weeks 1 to 6 per Cx43-wt FL were, respectively, 13 738, 5523, 2648, 853, 359, and 205, and for Cx43-KO FL they were, respectively, 10 148, 3369, 2061, 591, 316, and 195. (B) Colony-forming-cell content. Actual mean numbers were 3745 CFU-GM per Cx43-wt FL and 2876 per Cx43-KO FL, and 2215 BFU-E per Cx43-wt FL and 1525 BFU-E per Cx43-KO FL. *P < .05, between progenitor content of Cx43-wt and Cx43-KO FL cells.

Fig. 5.

Analysis of hemopoietic progenitor FL content from 14.5- to 15-day FLs from Cx43-wt (empty squares and bars) and Cx43-KO (filled squares and bars).

The results represent 3 independent experiments in which Cx43-wt (n = 11 fetuses; median: 7) and Cx43-KO (n = 9 fetuses; median: 3) FL cells had been obtained and compared within every experiment. (A) Weekly CAFC content. Actual mean numbers for weeks 1 to 6 per Cx43-wt FL were, respectively, 13 738, 5523, 2648, 853, 359, and 205, and for Cx43-KO FL they were, respectively, 10 148, 3369, 2061, 591, 316, and 195. (B) Colony-forming-cell content. Actual mean numbers were 3745 CFU-GM per Cx43-wt FL and 2876 per Cx43-KO FL, and 2215 BFU-E per Cx43-wt FL and 1525 BFU-E per Cx43-KO FL. *P < .05, between progenitor content of Cx43-wt and Cx43-KO FL cells.

Close modal

Effect of stromal Cx43 expression on their hemopoietic support ability

As the reduced content of hematopoietic progenitors in the Cx43-KO FL might be due to a hemopoietic defect, or alternatively, to an indirect effect on the animal development, we analyzed whether hemopoiesis, in vitro, was similarly affected by the level of stromal Cx43 expression. To eliminate possible variables between stromal cell lines derived from Cx43-wt and Cx43-KO mice that might affect their hemopoietic support ability, we reintroduced the Cx43 gene (FLB6-3 and FLB6-4 cell lines) into the Cx43 cell line (FLB6-2). As a source of hematopoietic progenitors, we used both normal and post–5-FU murine BM cells. The CAFC-week 1 support of normal BM cells was increased 369% in the FLB6-3 cells (P < .05) as compared with the Cx43 cell line (FLB6-2; Figure6A). The effect of Cx43 expression was clearer when coculturing stromal cells with 5-FU–treated BM, which is relatively enriched for primitive hematopoietic progenitors (Figure6B). As compared with Cx43 (FLB6-2) support, the reintroduction of Cx43 induced an increase of the CAFC frequency ranging from 194% (week 1) to 504% (week 4) when assayed on FLB6-3 cells and 307% (week 3) and 254% (week 4) when assayed on FLB6-4 cells (Figure 6B). CA support by FLB6-3 was significantly higher on weeks 1 and 2 than when assayed on FLB6-4 stromal cells. FLB6-3 cells, whose expression of Cx43 was 2.6-fold higher than FLB6-4, supported the formation of CAs, as an average, 4.3-fold better than FLB6-4 cells when analyzed on normal BM cells and 1.75-fold better than FLB6-4 when analyzed on 5-FU–treated BM cells. These results show that the level of membrane-bound Cx43 expression by stromal cells may correlate with their CAFC support ability in vitro.

Fig. 6.

Frequency of various CAFC-week types after coculture with different stromal cell lines.

FLB6-2 (Cx43) is represented by circles; fetal liver-derived FLB6-3 (Cx43+), by squares and FLB6-4 (Cx43+), by triangles. (A) When normal mouse BM cells were used for the coculture, the mean CAFC frequencies (per 105 cells) for weeks 1 to 4 were 36.06, 17.10, 2.10, and 1.05 (with the cell line FLB6-2); 40.0, 13.8, 1.7, and 0.8 (with the cell line FLB6-4); 61.7, 17.5, 2.8, and 1.1 (with the cell line FLB6-3). (B) When 5-FU–treated mouse BM cells were cocultured, the mean CAFC frequencies (per 105 cells) were 29.3, 28.7, 3.1, and 0.5 (with the cell line FLB6-2); 32.7, 30.6, 9.8, and 1.3 (with the cell line FLB6-4); and 54.9, 17.1, 8.0, and 3.1 (with the cell line FLB6-3). Four experiments run in parallel were carried out for the different stromal cell lines except for the analysis of CAFC frequency of normal BM on the cell line FLB6-4 (n = 2). *P < .05 between CAFC support by FLB6-2 and FLB6-3, #P < <.05 between CAFC support by FLB6-2 and FLB6-4, ^P < .05 between CAFC support by FLB3 and FLB6-4.

Fig. 6.

Frequency of various CAFC-week types after coculture with different stromal cell lines.

FLB6-2 (Cx43) is represented by circles; fetal liver-derived FLB6-3 (Cx43+), by squares and FLB6-4 (Cx43+), by triangles. (A) When normal mouse BM cells were used for the coculture, the mean CAFC frequencies (per 105 cells) for weeks 1 to 4 were 36.06, 17.10, 2.10, and 1.05 (with the cell line FLB6-2); 40.0, 13.8, 1.7, and 0.8 (with the cell line FLB6-4); 61.7, 17.5, 2.8, and 1.1 (with the cell line FLB6-3). (B) When 5-FU–treated mouse BM cells were cocultured, the mean CAFC frequencies (per 105 cells) were 29.3, 28.7, 3.1, and 0.5 (with the cell line FLB6-2); 32.7, 30.6, 9.8, and 1.3 (with the cell line FLB6-4); and 54.9, 17.1, 8.0, and 3.1 (with the cell line FLB6-3). Four experiments run in parallel were carried out for the different stromal cell lines except for the analysis of CAFC frequency of normal BM on the cell line FLB6-4 (n = 2). *P < .05 between CAFC support by FLB6-2 and FLB6-3, #P < <.05 between CAFC support by FLB6-2 and FLB6-4, ^P < .05 between CAFC support by FLB3 and FLB6-4.

Close modal

The existence of GJ-like structures in hemopoietic tissues has been known for 20 years.28-30 It is known that GJ-IC can only be achieved by the expression of compatible Cxs.31 The existence of functional heteromeric channels (different Cxs within the same hemiconnexon) in different tissues32-34 and the unique ionic and size selectivities that are determined by each combination of Cxs35 make IC highly specific and regulated. Although some Cx mutations are associated with human diseases3,6 and eight different KO models in rodents have been described,13 36-41 no hematological impairment has been reported related to a defect in one single Cx gene. We show here that various Cxs are expressed by hemopoietic tissues and that the deletion of the Cx43 gene leads to measurable hemopoietic defects in vivo and in vitro.

It has been reported that hemopoietic-supporting BM stromal cells express Cx43 but not Cx26 or Cx32.42 The expression of other Cxs in them has not been studied, and, as far as we know, Cx expression in hemopoietic-supporting FL stromal cell lines has not been reported. Previous reports7,8 have suggested that Cx43 is solely, or predominantly, responsible for GJ-IC in stromal cells of the BM. By using a sensitive RT-PCR technique, we observed that in addition to Cx43, 4 other Cxs are expressed in FL stromal cells (ie, Cx30.3, Cx31, Cx31.1, and Cx45) and 2 other Cxs in BM stromal cells (ie, Cx31 and Cx45). Cx43 and Cx45 are widespread expressed Cxs but Cx31 has been preferentially found in keratinocytes20 and trophectoderm cells,43 whereas Cx30.3 and Cx31.1 have been described to be expressed in keratinocytes44 only. We show that all stromal cell lines analyzed express Cx45 mRNA, and Cx45 protein was localized at the contacting cell membrane surface. Cx31 mRNA was also expressed by almost all of the stromal cell lines, and we showed its membrane protein expression in either wt or Cx43-deficient FL stromal cells.

It has been reported that the conductance of Cx43-dependent GJs is much higher than that observed in Cx45 and other Cxs, and, therefore, negatively charged molecules could more easily pass through Cx43-dependent GJs,45 suggesting differential permeability properties among the different Cxs. Our observation that at least 3 Cxs are expressed in BM stromal cells and at least 5 in FL stromal cells suggests that GJ-IC among stromal cells could be a complex communication system with heteromeric and heterotypic combinations between the Cx proteins. Heterotypic coupling (different Cxs in each hemiconnexon) between Cx43 and Cx45 has been observed in HeLa cells transfected with different mouse Cxs. The possibility that Cx43 and Cx45 may also form heterotypic junctions in stromal cells from hemopoietic organs, therefore, exists. Heterotypic coupling of Cx31 with either Cx43 or Cx45 probably occurs at a far lower frequency.35 The ability of Cx30.3 and Cx31.1 to form heterotypic GJs has not been described, but, if they existed, their expression would significantly increase the chance of Cx combinations.

The extent to which cells are functionally coupled by GJ channels depends on many different control mechanisms (gene transcription, messenger stability, translation, posttranslational modifications, topological modifications, and assembly and/or removal from the cell membrane46). We were interested in finding a method that showed the widest possible range of types of IC. Some dyes show different permeability, depending on the type of Cx-forming GJs.35,47 LY dye transfer48 and transfer of loaded calcein18,49-51 techniques have been shown as methods of determining GJ-IC. LY dye transfer has been shown in osteoblasts as highly dependent on Cx43-GJ.10 In Cx45+/Cx43-KO fetal fibroblasts that were communicating (as assessed by double whole cell patch-clamp), LY dye transfer was completely abolished, which suggests that LY is at least less transported through Cx45 GJs.12 We show that calcein dye transfer is impaired in a Cx43-deficient FL cell line as compared with Cx43-wt FL cell lines but not abolished. The reintroduction of the Cx43 gene allows the recovery of their lost IC ability. All of this is in agreement with our finding that other Cxs than Cx43 are expressed and all or some of them are involved in the IC.

A latent network of Cx43-GJ has been proposed in normal quiescent marrow that can be up-regulated in neonatal marrow or after forced stem cell division.8 However, it is unknown whether Cx43 gene deletion causes hemopoietic effects. Mice lacking Cx43 die between day 16 of pregnancy and birth because of a cardiac malformation at the junction between right ventricle and outflow tract,13,16and, as an external anatomical detail, they show a swollen neck. These mice have also been described as having defects in their germ line and gonads,52 defects in neural crest cell migration,53 modifications to lenses,54impairment of heart ventricular conduction,55 and changes in intracellular calcium concentration after neuronal mechanical stimulation.56 

We describe, here, for the first time, that Cx43 deficiency impairs hemopoiesis. Cx43-KO day 14.5-15 fetuses have a lower content of progenitor and stem cells in their liver as compared with their wt littermates. Rather than this being an indirect effect of a possible developmental retardation, we show that this hemopoietic defect in Cx43-KO mice may result from an indirect effect of the Cx43 deletion in hemopoietic stromal cells, as we found a significant increase in the apparent CAFC outgrowth on Cx43 stromal cell lines with a reintroduced Cx43 gene. We previously showed that the blockade of all GJ-IC by amphotericin B completelyand reversibly inhibited the CA formation and hemopoiesis in stroma-contact cultures but not in stroma noncontact cultures, while also leaving the primary colony formation unaffected.9Specifically, a dramatic increase was observed on the number of late developing CAs when 5-FU–treated BM cells were cultured on Cx43 reintroduced stromal cell lines. Rosendaal et al8 showed an up-regulation of Cx43-dependent GJs in 5-FU–treated BM that peaked after 4-6 days after 5-FU treatment. Our finding that day 6 post–5-FU cells showed a higher increased support for apparent CA formation on Cx43-KO stromal cells after the reintroduction of Cx43 than on Cx43-KO cells may support the role of Cx43 in regenerating BM. A Cx31-KO model, recently described,36 may allow for the same type of studies of BM or FL stromal hemopoiesis support to analyze the independent contribution of other Cxs expressed in hemopoiesis-supporting stromal cells.

From our data, it is evident that mutual stromal cells may communicate via GJ-IC. However, it remains unclear whether stromal cells and hematopoietic (stem) cells use GJs to exchange molecules. The existence of in vitro functional GJs between stromal and BM leukocytes has been described as rare or not existing at all.7,42 57-59 We found no Cx expression by RT-PCR (Cx43, Cx45, or Cx31) in different sorted BM-leukocyte populations representing B and T lymphocytes, myeloid (monocytic-macrophagic and granulocytic), erythroblastic, and partly enriched hematopoietic progenitors (unpublished observations). All of these data make us believe that the Cx43 deficiency-related effects reported in this paper are mostly a consequence of a defect in the communication among stromal cells.

Together, these observations suggest that stromal functional Cx43-dependent GJ contribute to the stromal regulation of the clonal outgrowth of hematopoietic progenitors. It is likely that other Cxs contribute to stromal regulation of hemopoiesis, as we have previously reported that blockade of the total GJ system in stroma-supported cultures fully and reversibly blocks hemopoiesis.

Supported by a fellowship grant (J.A.C.) from Fundacion Areces, Madrid, Spain.

Reprints:Rob E. Ploemacher, Dept of Hematology, Faculty of Medicine, Erasmus University, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands; e-mail: ploemacher@hema.fgg.eur.nl.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Williams
DA
Stem cell model of hematopoiesis.
Hematology: Basic Principles and Practice.
2nd ed.
Hoffman
R
et al
1995
180
Churchill Livingstone
New York, NY
2
Long
MW
Blood cell cytoadhesion molecules.
Exp Hematol.
20
1992
288
301
3
Kumar
NM
Gilula
NB
The gap junction communication channel.
Cell.
84
1996
381
388
4
Goodenough
DA
Goliger
JA
Paul
DL
Connexins, connexons, and intercellular communication.
Annu Rev Biochem.
65
1996
475
502
5
Sohl
G
Degen
J
Teubner
B
Willecke
K
The murine gap junction gene connexin36 is highly expressed in mouse retina and regulated during brain development.
FEBS Lett.
428
1998
27
31
6
Spray
DC
Gap junctions proteins: where they live and how they die.
Circ Res.
83
1998
679
681
7
Dorshkind
K
Green
L
Godwin
A
Fletcher
WH
Connexin-43-type gap junctions mediate communication between bone marrow stromal cells.
Blood.
82
1993
38
45
8
Rosendaal
M
Green
CR
Rahman
A
Morgan
D
Up-regulation of the connexin43+ gap junction network in haemopoietic tissue before the growth of stem cells.
J Cell Sci.
107
1994
29
37
9
Rosendaal
M
Mayen
A
de Koning
A
Dunina-Barkovskaya
T
Krenacs
T
Ploemacher
R
Does transmembrane communication through gap junctions enable stem cells to overcome stromal inhibition?
Leukemia.
11
1997
1281
1289
10
Steinberg
TH
Civitelli
R
Geist
ST
et al
Connexin43 and connexin45 form gap junctions with different molecular permeabilities in osteoblastic cells.
EMBO J.
13
1994
744
750
11
Koval
M
Geist
ST
Westphale
EM
et al
Transfected connexin45 alters gap junction permeability in cells expressing endogenous connexin43.
J Cell Biol.
130
1995
987
995
12
Martyn
KD
Kurata
WE
Warn-Cramer
BJ
Burt
JM
TenBroek
E
Lau
AF
Immortalized connexin43 knockout cell lines display a subset of biological properties associated with the transformed phenotype.
Cell Growth Differ.
8
1997
1015
1027
13
Reaume
AG
de Sousa
PA
Kulkarni
S
et al
Cardiac malformation in neonatal mice lacking connexin 43.
Science.
267
1995
1831
1834
14
Slieker
WA
van der Loo
JC
de Rijk-de Bruijn
MF
Godrey
DI
Leenen
PJM
van Ewijk
W
ER-MP12 antigen, a new cell surface marker on mouse bone marrow cells with thymus-repopulating ability. II. Thymus-homing ability and phenotypic characterization of ER-MP12-positive bone marrow cells.
Int Immunol.
5
1993
1099
1107
15
Breems
DA
Blokland
EA
Neben
S
Ploemacher
RE
Frequency analysis of human primitive haematopoietic stem cell subsets using a cobblestone area forming cell assay.
Leukemia.
8
1994
1095
1104
16
Ya
J
Erdtsieck-Ernste
EB
de Boer
PA
et al
Heart defects in connexin43-deficient mice.
Circ Res.
82
1998
360
366
17
Beyer
EC
Paul
DL
Goodenough
DA
Connexin43: a protein from rat heart homologous to a gap junction protein from liver.
J Cell Biol.
105
1987
2621
2629
18
Ziambaras
K
Lecanda
F
Steinberg
TH
Civitelli
R
Cyclic stretch enhances gap junctional communication between osteoblastic cells.
J Bone Miner Res.
13
1998
218
228
19
Hennemann
H
Schwarz
HJ
Willecke
K
Characterization of gap junction genes expressed in F9 embryonic carcinoma cells: molecular cloning of mouse connexin31 and -45 cDNAs.
Eur J Cell Biol.
57
1992
51
58
20
Butterweck
A
Elfgang
C
Willecke
K
Traub
O
Differential expression of the gap junction proteins connexin45, -43, -40, -31, and -26 in mouse skin.
Eur J Cell Biol.
65
1994
152
163
21
Sambrook
J
Fritsch
EF
Maniatis
T
Molecular Cloning. A Laboratory Manual.
2nd edition.
1989
Cold Spring Harbor Laboratory Press
Plainview, N Y
22
Fiorenza
MT
Mangia
F
Quantitative RT-PCR amplification of RNA in single mouse oocytes and preimplantation embryos.
Biotechniques.
24
1998
618
623
23
Davies
TC
Barr
KJ
Jones
DH
Zhu
D
Kidder
GM
Multiple members of the connexin gene family participate in preimplantation development of the mouse.
Dev Genet.
18
1996
234
243
24
Ploemacher
RE
van der Sluijs
JP
Voerman
JS
Brons
NH
An in vitro limiting-dilution assay of long-term repopulating hematopoietic stem cells in the mouse.
Blood.
74
1989
2755
2763
25
Umezawa
A
Harigaya
K
Abe
H
Watanabe
Y
Gap-junctional communication of bone marrow stromal cells is resistant to irradiation in vitro.
Exp Hematol.
18
1990
1002
1007
26
Ploemacher
RE
van der Sluijs
JP
van Beurden
CA
Baert
MR
Chan
PL
Use of limiting-dilution type long-term marrow cultures in frequency analysis of marrow-repopulating and spleen colony-forming hematopoietic stem cells in the mouse.
Blood.
78
1991
2527
2533
27
Crow
DS
Beuer
EC
Paul
DL
Kobe
SS
Lau
AF
Phosphorylation of connexin 43 gap junction protein in uninfected and Rous sarcoma virus-transformed mammalian fibroblasts.
Mol Cell Biol.
10
1990
1754
1763
28
Campbell
FR
Gap junctions between cells of bone marrow: an ultrastructural study using tannic acid.
Anat Rec.
196
1980
101
107
29
Yamazaki
K
Sl/Sld mice have an increased number of gap junctions in their bone marrow stromal cells.
Blood Cells.
13
1988
421
435
30
Yamazaki
K
Zacharov
Y
Simmons
PJ
Dexter
TM
Allen
TD
A comparative morphometric study on the ultrastructure of adherent cells in long-term bone marrow culture from normal and congenitally anemic mice.
Blood Cells.
15
1989
343
364
31
White
TW
Paul
DL
Goodenough
DA
Bruzzone
R
Functional analysis of selective interactions among rodent connexins.
Mol Biol Cell.
6
1995
459
470
32
Jiang
JX
Goodenough
DA
Heteromeric connexons in lens gap junction channels.
Proc Natl Acad Sci U S A.
93
1996
1287
1291
33
He
DS
Jiang
JX
Taffet
SM
Burt
JM
Formation of heteromeric gap junction channels by connexins 40 and 43 in vascular smooth muscle cells.
Proc Natl Acad Sci U S A.
96
1999
6495
6500
34
Stauffer
KA
The gap junction proteins beta 1-connexin (connexin-32) and beta 2-connexin (connexin-26) can form heteromeric hemichannels.
J Biol Chem.
270
1995
6768
6772
35
Elfgang
C
Eckert
R
Lichtenberg-Frate
H
et al
Specific permeability and selective formation of gap junction channels in connexin-transfected HeLa cells.
J Cell Biol.
129
1995
805
817
36
Willecke
K
Kirchhoff
S
Plum
A
Temme
A
Thonnissen
E
Ott
T
Biological functions of connexin genes revealed by human genetic defects, dominant negative approaches and targeted deletions in the mouse.
Novartis Found Symp.
219
1999
76
88
37
Simon
AM
Goodenough
DA
Li
E
Paul
DL
Female infertility in mice lacking connexin 37.
Nature.
385
1997
525
529
38
Kirchhoff
S
Nelles
E
Hagendorff
A
Kruger
O
Traub
O
Willecke
K
Reduced cardiac conduction velocity and predisposition to arrhythmias in connexin40-deficient mice.
Curr Biol.
8
1998
299
302
39
Nelles
E
Butzler
C
Jung
D
et al
Defective propagation of signals generated by sympathetic nerve stimulation in the liver of connexin32-deficient mice.
Proc Natl Acad Sci U S A.
93
1996
9565
9570
40
Gabriel
HD
Jung
D
Butzler
C
et al
Transplacental uptake of glucose is decreased in embryonic lethal connexin26-deficient mice.
J Cell Biol.
140
1998
1453
1461
41
Simon
AM
Goodenough
DA
Diverse functions of vertebrate gap junctions.
Trends Cell Biol.
8
1988
477
483
42
Krenacs
T
Rosendaal
M
Connexin 43 gap junctions in normal, regenerating and culture mouse bone marrow and in human leukemias.
Am J Pathol.
152
1998
993
1004
43
Dahl
E
Winterhager
E
Reuss
B
Traub
O
Butterweck
A
Willecke
K
Expression of the gap junction proteins connexin31 and connexin43 correlates with communication compartments in extraembryonic tissues and in the gastrulating mouse embryo, respectively.
J Cell Sci.
109
1996
191
197
44
Hennemann
H
Dahl
E
White
JB
et al
Two gap junction genes, connexin 31.1 and 30.3, are closely linked on mouse chromosome 4 and preferentially expressed in skin.
J Biol Chem.
267
1992
17225
17233
45
Veenstra
RD
Size and selectivity of gap junction channels formed from different connexins.
J Bioenerg Biomembr.
28
1996
327
337
46
Yeager
M
Gilula
NB
Membrane topology and quaternary structure of cardiac gap junction ion channels.
J Mol Biol.
223
1992
929
948
47
Kwak
BR
Jongsma
HJ
Selective inhibition of gap junction channel activity by synthetic peptides.
J Physiol.
516
1999
679
685
48
Stewart
WW
Functional connections between cells as revealed by dye-coupling with a highly fluorescent naphthalimide tracer.
Cell.
14
1978
741
759
49
Lecanda
F
Towler
DA
Ziambaras
K
et al
Gap junctional communication modulates gene expression in osteoblastic cells.
Mol Biol Cell.
9
1998
2249
2258
50
Frenzel
EM
Johnson
RG
Gap junction formation between cultured embryonic lens cells is inhibited by antibody to N-cadherin.
Dev Biol.
179
1996
1
16
51
Tomasetto
C
Neveu
MJ
Daley
J
Horan
PK
Sager
R
Specificity of gap junction communication among human mammary cells and connexin transfectants in culture.
J Cell Biol.
122
1993
157
167
52
Juneja
SC
Barr
KJ
Enders
GC
Kidder
GM
Defects in the germ line and gonads of mice lacking connexin 43.
Biol Reprod.
60
1999
1263
1270
53
Huang
GY
Cooper
ES
Waldo
K
Kirby
ML
Gilula
NB
Lo
CW
Gap junction-mediated cell-cell communication modulates mouse neural crest migration.
J Cell Biol.
143
1998
1725
1734
54
Gao
Y
Spray
DC
Structural changes in lenses of mice lacking the gap junction protein connexin43.
Invest Ophtalmol Vis Sci.
39
1998
1198
1209
55
Thomas
SA
Schuessler
RB
Berul
CI
et al
Disparate effects of deficient expression of connexin43 on atrial and ventricular conduction: evidence for chamber-specific molecular determinants of conduction.
Circulation.
97
1998
686
691
56
Naus
CC
Bechberger
JF
Zhang
Y
et al
Altered gap junctional communication, intercellular signaling, and growth in cultured astrocytes deficient in connexin43.
J Neurosci Res.
49
1997
528
540
57
Alves
LA
de Carvalho
AC
Savino
W
Gap junctions: a novel route for direct cell-cell communication in the immune system?
Immunol Today.
19
1998
269
275
58
Alves
LA
Coutinho-Silva
R
Persechini
PM
Spray
DC
Savino
W
Campos de Carvalho
AC
Are there functional gap junctions or junctional hemichannels in macrophages?
Blood.
88
1996
328
334
59
Rosendaal
M
Gregan
A
Green
CR
Direct cell-cell communication in the blood-forming system.
Tissue Cell.
23
1991
457
470
Sign in via your Institution