In addition to the well-recognized role in extracellular matrix remodeling, the tissue inhibitor of metalloproteinases-1 (TIMP-1) has been suggested to be involved in the regulation of numerous biologic functions, including cell proliferation and survival. We therefore hypothesized that TIMP-1 might be involved in the homeostatic regulation of HSCs, whose biologic behavior is the synthesis of both microenvironmental and intrinsic cues. We found that TIMP-1−/− mice have decreased BM cellularity and, consistent with this finding, TIMP-1−/− HSCs display reduced capability of long-term repopulation. Interestingly, the cell cycle distribution of TIMP-1−/− stem cells appears distorted, with a dysregulation at the level of the G1 phase. TIMP-1−/− HSCs also display increased levels of p57, p21, and p53, suggesting that TIMP-1 could be intrinsically involved in the regulation of HSC cycling dynamics. Of note, TIMP-1−/− HSCs present decreased levels of CD44 glycoprotein, whose expression has been proven to be controlled by p53, the master regulator of the G1/S transition. Our findings establish a role for TIMP-1 in regulating HSC function, suggesting a novel mechanism presiding over stem cell quiescence in the framework of the BM milieu.

The capability of HSCs to maintain the homeostasis of the hematopoietic system is the result of a finely tuned balance between self-renewal and differentiation. The mechanisms responsible for this balance comprise both intrinsic and extrinsic factors, whose crosstalk eventually dictates the fate of stem cells in the framework of the BM niche.1-3  Beside the well-established structural function, the dynamic network of interacting macromolecules that constitutes the extracellular matrix (ECM) represents one of the most powerful sources of extrinsic factors generated by the BM microenvironment.4  The intricate architecture created by these molecules not only guarantees protection and mechanical support to the stem cell pool but also plays an active role in regulating their behavior. By binding growth factors, regulating their bio-availability, and enabling the interaction with cell-surface receptors, ECM components have been shown to modulate a variety of cellular functions, such as proliferation, survival, and differentiation.5 

ECM dynamic remodeling is controlled by metalloproteinases (MMPs), a class of Zn++-dependent proteinases, such as collagenases, gelatinases, and stromelysins, that participate in the digestion of many ECM components, under both physiologic and pathologic conditions.6  The enzymatic activity of MMPs is counterbalanced by several natural inhibitors, including the tissue inhibitors of metalloproteinases (TIMPs).7  Both MMPs and TIMPs are expressed by hematopoietic and stromal cells8  and are decisive regulators of the crosstalk between these 2 cellular entities. The mammalian TIMP family comprises 4 highly conserved members that reversibly block MMP-dependent proteolysis by forming noncovalent 1:1 stoichiometric complexes. Alterations in the balance between the enzymatic activities of MMPs and TIMPs have been linked to developmental defects and are associated with specific tumor microenvironments.9 

Although TIMPs were initially described as mere inhibitors of MMPs, recent findings have offered a different perspective on their biologic role, unveiling their multifaceted nature.10,11  In addition to inhibiting MMPs, TIMP-1 has been proven to play MMP-independent, cytokine-like activities and to be involved in cell growth, angiogenesis, apoptosis, and migration.12,13  For instance, Nakajima et al14  recently found that TIMP-3 plays a role in recruiting HSCs into the cell cycle.

Despite intense investigation, the coexistence of MMP-dependent and -independent functions has hindered the thorough dissection of the signaling pathways activated by TIMP-1, leaving the interpretation of its different biologic effects controversial and difficult to reconcile. Liu et al15  described the ability of TIMP-1 to protect human breast epithelial cells from apoptosis through the focal adhesion kinase/PI3K and MAPK signaling pathway. A similar activity has been described in the erythroleukemic cell line UT-7, with activation of the JAK2/PI3K/Akt cascade.16  The mechanisms underlying the activation of the molecular pathways downstream of TIMP-1 are also a matter of debate. The tetraspanin receptor CD63 protein has been identified as putative cell-surface receptor for TIMP-1 in human breast epithelial cells.17  In this model, TIMP-1 promotes cell survival through the activation of a CD63/integrin complex on the membrane of MCF10A cells. However, according to Lambert et al,18  TIMP-1 would form at the cell surface a ternary complex with pro-MMP-9 and CD44, which would in turn activate the signaling cascade, preventing apoptotic death in UT-7 cells.

Here, we investigate the functional behavior of HSCs in a model of TIMP-1−/− mice, showing that the obliteration of TIMP-1 alters the cell-cycle dynamics of long-term HSCs (LT-HSCs), thus affecting their capability of repopulating recipients after transplantation. According to our model, TIMP-1 deficiency increases p53 levels, thus blocking the transition of HSCs from G1 to S, through the p53-dependent down-regulation of CD44. Our study highlights a novel biologic role of TIMP-1 in the regulation of the HSC compartment and provides a new interpretative tool for the molecular dissection of the crosstalk between HSCs and the environment within they reside.

Animals and cells

Wild-type (WT) C57Bl/6, CD45.1 mice and homozygous C57Bl/6, CD45.2 TIMP-1−/− mice were obtained from The Jackson Laboratory and used at 6-12 weeks of age for all experiments. Animals were maintained in pathogen-free conditions at Baylor College of Medicine. The hematopoietic progenitor cell line 32D was obtained from American Type Culture Collection and maintained in RPMI 1640 medium, supplemented with 10% FBS and rIL-3. All animal studies were approved by the Baylor College of Medicine Animal Care and Use Committee.

Retroviral transduction of BM progenitor cells

The enforced expression of TIMP-1 was induced in WT Sca-1+ hematopoietic progenitors by retroviral transduction with murine stem cell virus–internal ribosome entry site–enhanced green fluorescent protein (MSCV-IRES-eGFP) or MSCV-TIMP1-IRES-eGFP as previously described.19  After transduction, cells were transplanted into lethally irradiated C57BL/6, CD45.1 recipient mice (50 000 cells/mouse).

In vitro CFU assay

Side population+c-Kit+Sca-1+Lineage (SPKLS) HSCs or KSL cells were isolated from the BM of WT and TIMP-1−/− mice and sorted on a MoFlo cytometer into 96-well plates (1 cell/well), containing M3434 MethoCult medium (StemCell Technologies), and incubated at 37°C, 5% CO2. After 12-14 days of culture, colonies were counted. For in vitro CFU (CFU-C) of cells overexpressing TIMP-1, 32D cells were transduced with retroviral vectors (MSCV-IRES-eGFP or MSCV-TIMP1-IRES-eGFP) as described and cultured in transduction medium at 37°C, 5% CO2 for 48 hours. For primary cells, Sca-1+ cells were transduced as described.19  GFP+Sca-1+ cells were then sorted into 96-well plates and cultured as above.

Spleen CFU assay

BM cells (200 000) from C57Bl/6, CD45.2 WT or TIMP-1−/− mice were transplanted into C57Bl/6, CD45.1 lethally irradiated recipients. Thirteen days after transplantation, spleens were harvested, fixed in Bouin solution for 30 minutes, and spleen CFUs (CFU-Ss) were counted under a dissecting microscope. Similarly, Sca-1+ cells, obtained from C57Bl/6, CD45.2 mice treated with 5-fluorouracil, were retrovirally transduced as described with MSCV constructs, and 35 000 Sca-1+–transduced cells were transplanted into C57Bl/6, CD45.1 recipient mice. Spleens were fixed and CFU-Ss were scored as above.

Transplantation assays

BM cells from 3 mice (sex- and age-matched) were harvested from tibias and femurs of TIMP-1−/− and WT C57Bl/6, CD45.2 mice. Whole BM (WBM) cells (200 000) from each group were transplanted into primary C57Bl/6, CD45.1 recipients, along with an equal number of CD45.1 WBM competitor cells (1:1). Peripheral blood (PB) engraftment and lineage distribution of CD45.2 cells was analyzed periodically at monthly intervals. Repopulation units were calculated as per Harrison.20  A separate set of transplantations was performed to test the effects of a TIMP-1−/− microenvironment on engraftment. CD45.1 WT BM cells (400 000) were transplanted into C57Bl/6, CD45.2 WT, or TIMP-1−/− recipients. PB chimerism and lineage distribution of CD45.1 cells was analyzed periodically at 3, 6, and 9 weeks after transplantation.

Flow cytometry

PB from untreated or mice that received a transplant was collected from the retro-orbital sinus. The samples were pretreated with red blood cell lysis buffer (Tris Cl pH 7.8 and ammonium chloride, 1:9) and then stained for 20 minutes on ice with the following Ab cocktail: anti-CD45.2 (or CD45.1) APC-conjugated, anti-CD4 Pacific Blue, anti-CD8 Pacific Blue, anti-B220 Pacific Blue, anti-B220 PE-cyanine 7, anti-Mac1 PE-cyanine 7, and anti-Gr1 PE. Cells were resuspended in Hanks buffer containing propidium iodide (PI; 1 μg/mL). The analysis of common lymphoid progenitors (CLPs) and common myeloid progenitors (CMPs), as well as MMPs, short-term HSCs, and LT-HSCs, was performed as previously described.20  Samples were analyzed on an LSRII flow cytometer (Becton Dickinson).

Cell cycle

Cell cycle analysis was performed as previously described.21  Briefly, SPKLS HSCs22  were sorted on top of 200 000 B220+ splenocytes. Subsequently, cells were stained for 45 minutes at 37°C with Hoechst 33342 (20 μg/mL) and verapamil (50 μg/mL; Sigma-Aldrich) in PBS plus 3% FBS. Pyronin Y (1 μg/mL; Sigma-Aldrich) was then added to the staining solution, and cells were incubated for 15 minutes at 37°C. Ki-67 staining was performed as previously described.20  Samples were analyzed on an LSRII flow cytometer (Becton Dickinson).

Apoptosis

B220+ splenocytes (200 000) were sorted into collection tubes. Subsequently, SPKLS HSCs were sorted into the same tubes, spun down, and resuspended in Annexin Binding Buffer (10mM HEPES, 140mM NaCl, 2.5mM CaCl2, pH 7.4) and stained for 15 minutes at room temperature with anti-Annexin V antibody and PI. Samples were immediately analyzed on an LSRII flow cytometer (Becton Dickinson).

Migration assay

Transmigration assays were performed in 24-well transwell plates (diameter, 6.5 mm; pore size, 5 μm; Corning International). Sca-1+ cells (250 000) were loaded into the upper chamber in 100 μL of StemPro Medium, supplemented with nutrient supplement, L-glutamine, and penicillin/streptomycin. Lower chambers were loaded with 600 μL of the same media, supplemented with 125 ng/mL CXCL12 (R&D Systems). Plates were incubated at 37°C, 5% CO2 for 4 hours. Migration rate was calculated as follows: (migrated cells/loaded cells) × 100. CD150+48KSL migration rate was calculated on the basis of their frequency in the original population and in the migrated cells.

Real-time PCR analysis

SPKLS cells from WT and TIMP-1−/− mice (n = 3 per group) were sorted into lysis buffer for RNA, and real-time PCR was performed as previously described. The fold change was calculated from the formula 2ΔΔCt. For each gene the WT value was set to 1. The error bars represent the corresponding SEM.

Statistics

Statistical significance was determined by 2-tailed Student t test. P values are indicated as *P < .05, **P < .01, and ***P < .001.

Phenotypic characterization of TIMP-1−/− mice shows impairment of the HSC compartment

To begin to investigate the role of TIMP1 in regulation of HSCs, we first examined the hematopoietic system of TIMP-1−/− mice. We initially investigated whether the constitutive lack of TIMP-1 would affect the production and release of mature cells in the PB. TIMP-1−/− mice showed no major alteration in the respective percentages of circulating myeloid cells (Mac-1+), as well as B lymphocytes (B220+) and T lymphocytes (CD4+CD8+), when assessed on a flow cytometer (Figure 1A). Likewise, complete blood count analyses performed on knockout (KO) mice did not show any significant deviation from the PB profile of the WT counterpart (L.R., unpublished data). Similarly, the analysis of BM and spleens of KO mice did not show any relevant difference in the linage distribution of hematopoietic cells (Figure 1A; supplemental Figure 1A, available on the Blood Web site; see the Supplemental Materials link at the top of the online article).

Figure 1

Phenotypic characterization of hematopoiesis in TIMP-1−/− mice. (A) Lineage distribution of Mac-1+ myeloid cells, B220+ B lymphocytes, and CD4+CD8+ T lymphocytes in the BM, PB, and spleen of WT and TIMP-1−/− mice, as assessed by flow cytometry. (B) BM cellularity (absolute numbers) per mouse. Cells were harvested from both tibias, femurs, and hips. Results are reported as mean ± SEM.

Figure 1

Phenotypic characterization of hematopoiesis in TIMP-1−/− mice. (A) Lineage distribution of Mac-1+ myeloid cells, B220+ B lymphocytes, and CD4+CD8+ T lymphocytes in the BM, PB, and spleen of WT and TIMP-1−/− mice, as assessed by flow cytometry. (B) BM cellularity (absolute numbers) per mouse. Cells were harvested from both tibias, femurs, and hips. Results are reported as mean ± SEM.

Close modal

We next analyzed the BM compartment and surprisingly observed that TIMP-1−/− mice consistently presented with a decreased BM cellularity (average decrease of 24.8%; P < .001), compared with WT mice (Figure 1B). The decreased BM cellularity is not ascribable to a smaller size of the KO mice, because their weight in grams is comparable to that of WT mice (supplemental Figure 1B). Interestingly, we found that TIMP-1−/− mice also present a slightly decreased percentage of HSCs (Figure 2A), when quantified on the basis of the SPKSL phenotype. SPKSL absolute numbers also presented a 43% decrease, compared with controls (4.9 × 103 ± 0.7 × 103 and 8.6 × 103 ± 1.1 × 103, respectively; P < .05; Figure 2B). Similar results were obtained when HSC percentages and absolute numbers were quantified on the basis of the CD150+48KSL phenotype (Figure 2C-D; supplemental Figure 2). The CD34/Flk2 markers were also used to analyze HSC frequencies. With this particular staining scheme the percentage of LT-HSCs in TIMP-1−/− mice appeared to be significantly decreased compared with WT animals (P = .05; Figure 2E; supplemental Figure 1C-D).

Figure 2

Phenotypic characterization of TIMP-1−/− stem and progenitor cells. (A-B) Percentages and absolute numbers of BM SPKLS HSCs from WT and TIMP-1−/− mice. (C-D) Percentages and absolute numbers of BM CD150+48KSL HSCs from WT and TIMP-1−/− mice. (E) Absolute numbers of BM MMPs, short-term HSCs, and LT-HSCs. (F) Percentage of BM KSLs in WT and TIMP-1−/− mice. (G) Percentage of CLPs and common myeloid (granulocyte macrophage progenitors [GMPs], CMPs, and megakaryocyte-erythroid precursors [MEPs]) progenitors in WT and TIMP-1−/− mice. (H) Percentage of KSL cells in the spleen of WT and TIMP-1−/− mice. Results are reported as mean ± SEM of at least triplicate experiments. *P < .05 and ***P < .001.

Figure 2

Phenotypic characterization of TIMP-1−/− stem and progenitor cells. (A-B) Percentages and absolute numbers of BM SPKLS HSCs from WT and TIMP-1−/− mice. (C-D) Percentages and absolute numbers of BM CD150+48KSL HSCs from WT and TIMP-1−/− mice. (E) Absolute numbers of BM MMPs, short-term HSCs, and LT-HSCs. (F) Percentage of BM KSLs in WT and TIMP-1−/− mice. (G) Percentage of CLPs and common myeloid (granulocyte macrophage progenitors [GMPs], CMPs, and megakaryocyte-erythroid precursors [MEPs]) progenitors in WT and TIMP-1−/− mice. (H) Percentage of KSL cells in the spleen of WT and TIMP-1−/− mice. Results are reported as mean ± SEM of at least triplicate experiments. *P < .05 and ***P < .001.

Close modal

Conversely, the KSL compartment showed a slight increase compared with WT mice (Figure 2F), although the absolute number of KSLs still remains decreased. To determine whether the KSL compartment would present any specific skewing, we also analyzed the relative percentages of CMPs and CLPs. As assessed by flow cytometry, none of the myeloid progenitors (megakaryocyte-erythroid precursors, CMPs, and granulocyte macrophage progenitors), as well as the CLPs, seemed to be affected by TIMP-1 deficiency, suggesting that the slight expansion KSL cells is equally distributed among the different classes of progenitors (Figure 2G).

These data suggest that TIMP-1 deficiency affects the hematopoietic system, especially in terms of BM cellularity. Nonetheless, the production of mature PB components seems not to be significantly affected. We therefore hypothesized the existence of compensatory mechanisms promoted by extramedullary hematopoietic sites, such as the spleen. We found that the percentage of KSL progenitors in TIMP-1−/− spleens was significantly higher than the controls (1.5-fold increase; P < .05; Figure 2H), suggesting a pivotal role of splenic hematopoiesis in rescuing the PB phenotype.

Hematopoietic compartment in TIMP-1−/− mice is functionally impaired

In an attempt to investigate how the described phenotype reflects on BM functionality, we first assessed the clonogenic potential of BM-derived SPKLS cells in vitro. The number of colonies generated in semisolid cultures after 10 and 14 days was significantly increased (1.4-fold at day 10, P < .001; 1.2-fold at day 14; P < .05; Figure 3A) compared with the controls. However, at day 18, although the number of WT colonies kept increasing, the number of CFU-Cs generated from TIMP-1–deficient SPKLS cells remained the same. Interestingly, we noticed that the size of the colonies generated from TIMP-1−/−SPKLS cells was also enlarged compared with the WT (data not shown). Conversely, KSL cells did not display any significant difference in terms of clonogenic potential (supplemental Figure 3), when cultured in the same conditions.

Figure 3

Functional characterization of hematopoiesis in TIMP-1−/− mice. (A) Clonogenic assay of BM SPKLS cells isolated from WT and TIMP-1−/− mice and sorted as single cells in M3434 MethoCult. CFU-Cs were macroscopically scored at 10, 14, and 18 days after seeding. (B) Spleen-colony assay performed by transplanting 200 000 WBM cells (WT or TIMP-1−/−) into WT recipients. CFI-S formation was assessed by splenectomy 13 days after transplantation. (C) Schematic representation of the MSCV retroviral expression vectors used to transduce Sca-1+ hematopoietic progenitors. The control construct (top) only expresses the reporter gene GFP, whereas the TIMP-1 construct (bottom) allows the coordinated expression of both TIMP-1 and GFP. (D) Proliferation of 32D cells in liquid culture after transduction with MSCV constructs (P < .05; left). (E) For CFU-C assessment, 32D cells were transduced with MSCV vectors and sorted as single cells, after 48 hours, into 96-well plates and grown in MethoCult for 14 days. (F) Primary Sca-1+ progenitors (50 000) were transduced with control or MSCV–TIMP-1 constructs and transplanted into recipient mice. CFU-S colonies were analyzed by splenectomy on day 13 after transplantation. Results are reported as mean ± SEM of at least triplicate experiments. *P < .05 and ***P < .001.

Figure 3

Functional characterization of hematopoiesis in TIMP-1−/− mice. (A) Clonogenic assay of BM SPKLS cells isolated from WT and TIMP-1−/− mice and sorted as single cells in M3434 MethoCult. CFU-Cs were macroscopically scored at 10, 14, and 18 days after seeding. (B) Spleen-colony assay performed by transplanting 200 000 WBM cells (WT or TIMP-1−/−) into WT recipients. CFI-S formation was assessed by splenectomy 13 days after transplantation. (C) Schematic representation of the MSCV retroviral expression vectors used to transduce Sca-1+ hematopoietic progenitors. The control construct (top) only expresses the reporter gene GFP, whereas the TIMP-1 construct (bottom) allows the coordinated expression of both TIMP-1 and GFP. (D) Proliferation of 32D cells in liquid culture after transduction with MSCV constructs (P < .05; left). (E) For CFU-C assessment, 32D cells were transduced with MSCV vectors and sorted as single cells, after 48 hours, into 96-well plates and grown in MethoCult for 14 days. (F) Primary Sca-1+ progenitors (50 000) were transduced with control or MSCV–TIMP-1 constructs and transplanted into recipient mice. CFU-S colonies were analyzed by splenectomy on day 13 after transplantation. Results are reported as mean ± SEM of at least triplicate experiments. *P < .05 and ***P < .001.

Close modal

In addition, BM cells from TIMP-1−/− mice were transplanted into lethally irradiated mice, and the appearance of spleen colonies (CFU-S) was quantified. Thirteen days after transplantation, the total number of CFU-Ss generated by TIMP-1−/− BM cells was considerably expanded with respect to the WT (14.0 and 9.2 CFU-Ss, respectively; P < .05; Figure 3B).

To further investigate the role of TIMP-1 in cell proliferation, we transduced 32D cells with retroviral vectors driving the coordinated expression of TIMP-1 and eGFP (Figure 3C). We found that the enforced expression of TIMP-1 reduced cell proliferation of 32D cells in liquid culture (42.3% decrease; P < .05; Figure 3D) and significantly suppressed the generation of CFU-Cs in vivo (P < .05; Figure 3E). With the use of the same retroviral vectors, we also transduced primary WT Sca-1+ hematopoietic progenitor cells/HSCs and tested how the overexpression of TIMP-1 would affect CFU-S generation after transplantation. We found the CFU-S output to be significantly decreased after the induction of TIMP-1–enforced expression (1.8 fold-decrease; P < .001; Figure 3F). These findings reinforce the supposition that TIMP-1 plays a role in controlling proliferative dynamics.

Long-term hematopoietic reconstitution sustained by TIMP-1−/− HSCs is compromised

We subsequently evaluated the capability of TIMP-1−/− HSCs to replenish the hematopoiesis of lethally irradiated recipients, by inducing long-term, multilineage reconstitution on transplantation. WBM cells were transplanted along with WT competitors (1:1), and their engraftment was periodically evaluated as contribution to PB chimerism (Figure 4A). Under these conditions, we observed that TIMP-1−/− BM cells do not reconstitute recipients as efficiently as WT cells (21.9% vs 53.3%; P < .05; 12 weeks after transplantation; Figure 4B). Of note, a profound decrease of 5.9-fold in repopulation units was observed in primary recipients of TIMP-1−/− WBM transplants (Figure 4C). We also analyzed the KSL compartment of mice that received a transplant for contribution from donor-derived cells, as measured by expression of CD45.2 Ag. We found that, 12 weeks after transplantation, mice that received a transplant with WT BM cells present a 12.2% contribution of donor-derived BM KSLs, whereas in mice receiving TIMP-1 KO WBM cells, the contribution to the KSL compartment drastically drops to 4.8% (Figure 4D; P = .0005). Distribution among the different hematopoietic lineages was also evaluated, although no significant differences were observed compared with the control (L.R., unpublished data). These findings indicate that, despite the ability of TIMP-1−/− BM cells to sustain multilineage reconstitution, their long-term reconstitution is significantly affected when in direct competition with their WT counterparts.

Figure 4

Competitive transplantation of TIMP-1−/− BM cells into WT recipients. (A) Schematic representation of the experimental model used for competitive transplantations of WT or TIMP-1−/− BM cells into WT recipients. (B) Percentage of engraftment at 4, 8, and 12 weeks after transplantation, calculated as relative PB chimerism of donor CD45.2 cells. (C) Repopulation units at 12 weeks after transplantation, calculated according to Harrison20  (see “Transplantation assays”). (D) Percentage of donor-derived (CD45.2) progenitors in the BM KSL compartment at 12 weeks after transplantation. Results are reported as mean ± SEM. *P < .05 and ***P < .001.

Figure 4

Competitive transplantation of TIMP-1−/− BM cells into WT recipients. (A) Schematic representation of the experimental model used for competitive transplantations of WT or TIMP-1−/− BM cells into WT recipients. (B) Percentage of engraftment at 4, 8, and 12 weeks after transplantation, calculated as relative PB chimerism of donor CD45.2 cells. (C) Repopulation units at 12 weeks after transplantation, calculated according to Harrison20  (see “Transplantation assays”). (D) Percentage of donor-derived (CD45.2) progenitors in the BM KSL compartment at 12 weeks after transplantation. Results are reported as mean ± SEM. *P < .05 and ***P < .001.

Close modal

TIMP-1 defect is cell autonomous and is intrinsic to HSCs rather than the BM microenvironment

HSC function and survival highly rely on interactions with the hematopoietic niche. Because of the role that TIMP-1 plays in tuning MMP function, we considered that the impaired repopulation activity of TIMP-1−/− BM cells might be secondary to alterations at the interface between HSCs and the niche. To address this possibility, we transplanted WT WBM cells into WT or TIMP-1−/− recipients and periodically evaluated their engraftment and lineage distribution (Figure 5A). Over a period of 9 weeks, we found that the percentage of donor-derived chimerism in the PB was essentially the same in the 2 groups (88.1% and 83.9% for TIMP-1 KO mice and WT mice, respectively; Figure 5B), suggesting that the capability of WT BM cells of repopulating lethally irradiated mice is not significantly affected by the constitutive lack of TIMP-1 in the BM niche. The lineage distribution of PB cells also did not appear to significantly diverge from the control (Figure 5C), although B-cell production appeared to be slightly decreased at the latest time point. Because of the role played by MMPs/TIMPs in regulating HSC trafficking, we hypothesized that TIMP-1 deficiency may determine a decreased BM cellularity by increasing HSC mobilization into the PB. However, the percentage of circulating KSLs in KO mice did not appear to be significantly different from the controls (Figure 5D). Furthermore, compared with WT controls, TIMP-1−/− cells showed the same homing rate toward the BM or the spleen (L.R., unpublished data), suggesting that the hematopoietic impairment observed in TIMP-1−/− mice most probably depends on a defect intrinsic to the HSCs, rather than their interaction with the BM environment.

Figure 5

Microenvironmental effects TIMP-1 deficiency on engraftment. (A) Schematic representation of the experimental model used, consisting of transplanting WT BM cells into either WT or TIMP-1−/− animals. (B) Percentage of engraftment at 3, 6, and 9 weeks after transplantation, calculated as relative PB chimerism of donor CD45.1 cells. (C) Lineage distribution of Mac-1+ myeloid cells, B220+ B lymphocytes, and CD4+CD8+ T lymphocytes in the PB of animals that received a transplant at 6 weeks. Results are percentages of the donor-derived white cell CD45.1 subset. (D) Percentages of circulating KLS cells in the PB of WT and TIMP-1−/− mice. Results are reported as mean ± SEM.

Figure 5

Microenvironmental effects TIMP-1 deficiency on engraftment. (A) Schematic representation of the experimental model used, consisting of transplanting WT BM cells into either WT or TIMP-1−/− animals. (B) Percentage of engraftment at 3, 6, and 9 weeks after transplantation, calculated as relative PB chimerism of donor CD45.1 cells. (C) Lineage distribution of Mac-1+ myeloid cells, B220+ B lymphocytes, and CD4+CD8+ T lymphocytes in the PB of animals that received a transplant at 6 weeks. Results are percentages of the donor-derived white cell CD45.1 subset. (D) Percentages of circulating KLS cells in the PB of WT and TIMP-1−/− mice. Results are reported as mean ± SEM.

Close modal

Cell cycle distribution and completion are altered in TIMP-1−/− HSCs

The decreased BM cellularity and the impaired BM repopulation after transplantation led us to hypothesize that TIMP-1 deficiency may be responsible for cell-cycle deregulation in HSCs. We used Pyronin Y staining in conjunction with Hoechst, that distinguishes cells in the G0 (PyroninY/Hoechst), G1 (PyroninY+/Hoechst), and S-G2M (Hoechst+) phases (Figure 6A).21,23  In agreement with our hypothesis, we found that the cell cycle distribution of TIMP-1−/− SPKLS cells was significantly deregulated (Figure 6B). As shown in Figure 6B, the percentages of SPKLS cells in the G0 phase of the cell cycle were decreased in TIMP-1−/− mice compared with the control (79.5% ± 3.5% vs 87.6% ± 2.3%, respectively; P < .01), whereas a 2.6-fold increase in the percentage of HSCs in the G1 phase was accordingly recorded (17.4% ± 3.2% for TIMP-1−/− SPKLS cells vs 6.8% ± 1.7% for WT SPKLS cells; P < .01). The percentage of TIMP-1−/− HSCs found to be in the S/G2/M phase also appeared decreased compared with the WT counterpart. Such a cell-cycle distribution suggests that TIMP-1−/− mice are depleted in long-term, quiescent HSCs (which are assumed to be mainly in G0). In addition, in the constitutive absence of TIMP-1, the physiologic cell-cycle completion appears to be impaired, because TIMP-1−/− HSCs present a dysregulation at the level of the G1-S transition. The role of TIMP-1 deficiency in HSC cell cycle was also investigated by measuring Ki-67, a nuclear marker of proliferation. TIMP-1−/− SPKLS cells express significantly higher levels of Ki-67 than the WT counterpart, thus confirming the finding that TIMP-1−/− mice have a lower percentage of G0 HSCs.

Figure 6

Cell proliferation and survival in TIMP-1−/− SPKLS HSCs. (A) Representative flow cytometric plots of cell-cycle analysis performed with PyroninY/Hoechst 33324 staining on WT or TIMP-1−/− SPKLS LT-HSCs. (B) Cell-cycle analysis of SPKLS HSCs and percentage distribution in the phases G0, G1, and S-G2M. The percentages of SPKLS in the G0 phase of the cell cycle were considerably decreased in TIMP-1−/− mice than in compared with the control (P < .05), whereas a 2.6-fold increase in the percentage of HSCs in the G1 phase was accordingly recorded (P < .05). (C) Percentages of Ki-67+ cells in WT and TIMP-1−/− SPKLS cells. (D) Cell survival analysis performed by annexin V staining on HSCs immediately after isolation. We observed that the percentage of Annexin V+ SPKLS cells was essentially the same in the 2 groups. (E) Expression levels of cell-cycle inhibitors in SPKLS cells was assessed by real-time PCR. Values are the average of duplicate experimental replicates. For each group, SPKLS cells from 3 different animals were pooled together for RNA extraction. *P < .05 and **P < .01.

Figure 6

Cell proliferation and survival in TIMP-1−/− SPKLS HSCs. (A) Representative flow cytometric plots of cell-cycle analysis performed with PyroninY/Hoechst 33324 staining on WT or TIMP-1−/− SPKLS LT-HSCs. (B) Cell-cycle analysis of SPKLS HSCs and percentage distribution in the phases G0, G1, and S-G2M. The percentages of SPKLS in the G0 phase of the cell cycle were considerably decreased in TIMP-1−/− mice than in compared with the control (P < .05), whereas a 2.6-fold increase in the percentage of HSCs in the G1 phase was accordingly recorded (P < .05). (C) Percentages of Ki-67+ cells in WT and TIMP-1−/− SPKLS cells. (D) Cell survival analysis performed by annexin V staining on HSCs immediately after isolation. We observed that the percentage of Annexin V+ SPKLS cells was essentially the same in the 2 groups. (E) Expression levels of cell-cycle inhibitors in SPKLS cells was assessed by real-time PCR. Values are the average of duplicate experimental replicates. For each group, SPKLS cells from 3 different animals were pooled together for RNA extraction. *P < .05 and **P < .01.

Close modal

Because TIMP-1 has previously been suggested to play a role in cell survival,16,18  we also considered the possibility that the impaired reconstitution ability of TIMP-1−/− BM cells could be secondary to a higher rate of cell death in the hematopoietic compartment. We addressed this possibility by comparing the percentage of CD150+48 KSL cells undergoing spontaneous apoptosis in TIMP-1−/− and WT mice, as measured by annexing V/PI staining. We observed that the percentage of annexin V+ SPKLS HSCs was essentially the same in the 2 groups, (Figure 6D; supplemental Figure 4). These data suggest that, at least under the conditions we investigated, HSC survival is not significantly affected by TIMP-1 deficiency.

To further investigate the role of TIMP-1 in HSC cell cycle, we analyzed by real-time PCR the expression level of some of the prominent regulators of cycling activity24,25  in SPKLS cells (Figure 6E). We found that p21 mRNA levels display a 3-fold increase in TIMP-1−/− SPKLS cells. Likewise, the expression levels of p57, found to play a pivotal role in HSC quiescence,26,27  showed a 4.7-fold increase in TIMP-1−/− SPKLS cells, whereas p27 levels in KO HSCs were only slightly decreased compared with WT HSCs (Figure 6E).

In vitro migration and expression of CD44 is significantly decreased in TIMP-1−/− LT-HSCs

Efficient homing and subsequent engraftment of HSCs into the BM microenvironment relies on 2-way molecular interactions between donor-derived hematopoietic cells and the recipient niche.28  Alterations in the ability of each of these counterparts to efficiently crosstalk could compromise the entrance of HSCs in the niche and their subsequent survival. Although we showed that a TIMP-1–deficient niche does not hinder the engraftment of WT BM cells (Figure 5), the capability of TIMP-1 HSCs per se to properly migrate toward the BM microenvironment still remained unclear. We therefore considered the possibility that the decreased repopulation observed for TIMP-1−/− BM cells could be ascribed to an impaired migratory pattern, in addition to the aforementioned proliferation defects. To address this possibility, we analyzed the capability of TIMP-1−/− progenitors of migrating toward a gradient of the chemokine CXCL12 (stromal cell-derived factor 1), which represents the most powerful regulator of HSC directional migration. The percentage of Sca-1+ hematopoietic progenitor cells migrating in vitro toward CXCL12 was significantly lower for TIMP-1−/− mice than for controls, with a 39% decrease in the migration rate (P < .05; Figure 7A). To define more precisely how TIMP-1 deficiency affects HSC migration, we calculated the percentage of CD150+48KSLs that underwent migration in both groups. Consistent with previous results, we observed that CD150+48KSLs from KO mice are less responsive to CXCL12 gradients and migrate at a significantly lower rate (P = .05; Figure 7B). Of note, we observed that the membrane expression of CXCR4, the conjugate receptor for CXCL12, is not significantly affected by TIMP-1 deficiency. When analyzed in a pool of BM CD150+48KSL HSCs, no difference in the MFI of CXCR4 was observed (Figure 7C; supplemental Figure 5), indicating that CXCR4 expression cannot account for the impaired migration of TIMP-1−/− HSCs.

Figure 7

Chemotactic migration of TIMP-1−/− HSCs toward CXCL12 and expression of CD44 and p53. (A-B) Percentage of chemotactic migration, toward a gradient of CXCL12, of Sca-1+ cells or CD150+48KSL HSCs from WT or TIMP-1−/− mice. Chemotactic assays were performed with transwell chambers, and cells were allowed to migrate for 4 hours at 37°C, 5% CO2. (C) MFI of CXCR4 expression on BM CD150+ 48KSL HSCs. (D-E) MFI of CD44 expression on the cell membrane of WT and TIMP-1−/− SPKLS HSCs. Results are reported as mean ± SEM of at least triplicate experiments; *P < .05. (F) p53 expression was measured in WT and TIMP-1−/− SPKLS cells by real-time PCR. Values are the average of duplicate experimental replicates. For each group, SPKLS cells from 3 different animals were pooled together for RNA extraction; *P < .05.

Figure 7

Chemotactic migration of TIMP-1−/− HSCs toward CXCL12 and expression of CD44 and p53. (A-B) Percentage of chemotactic migration, toward a gradient of CXCL12, of Sca-1+ cells or CD150+48KSL HSCs from WT or TIMP-1−/− mice. Chemotactic assays were performed with transwell chambers, and cells were allowed to migrate for 4 hours at 37°C, 5% CO2. (C) MFI of CXCR4 expression on BM CD150+ 48KSL HSCs. (D-E) MFI of CD44 expression on the cell membrane of WT and TIMP-1−/− SPKLS HSCs. Results are reported as mean ± SEM of at least triplicate experiments; *P < .05. (F) p53 expression was measured in WT and TIMP-1−/− SPKLS cells by real-time PCR. Values are the average of duplicate experimental replicates. For each group, SPKLS cells from 3 different animals were pooled together for RNA extraction; *P < .05.

Close modal

Because of the correlation between migratory behavior and expression of adhesion molecules, we also considered whether their expression in TIMP-1−/− HSCs would be affected. Interestingly, we found that the expression of CD44, a glycoprotein highly expressed on LT-HSCs,28-34  was significantly influenced by the constitutive lack of TIMP-1. When assessed by flow cytometry, CD44 MFI emerged to be decreased in TIMP-1−/− SPKLS HSCs, with a 14.5% reduction compared with the WT controls (P < .05; Figure 7D-E; supplemental Figure 6). Because of the role that CD44 has been suggested to play in both migration and proliferation,29  the finding that this glycoprotein is down-regulated in TIMP-1−/− HSCs might provide a possible explanation for their deficient responsiveness to CXCL12 gradients, as well as their aberrant cell cycle, which could ultimately be responsible for the decreased BM cellularity in TIMP-1–deficient mice. Interestingly, Godar et al35  showed that CD44 expression is directly inhibited by the tumor suppressor p53. By real-time PCR we measured p53 expression in SPKLS cells and found that p53 levels are 3.6 times higher in TIMP-1−/− HSCs compared than in controls (Figure 7F).

Although the role of TIMP-1 in ECM turnover has been extensively investigated, a thorough analysis of its MMP-independent, cytokine-like functions in hematopoiesis is yet to be provided. In this study we demonstrate that TIMP-1 subtly affects the HSC compartment, by subverting cell-cycle dynamics of long-term repopulating HSCs. BM cellularity in TIMP-1−/− mice is significantly decreased, with a concomitant reduction in the absolute number of LT-HSCs. When tested in a competitive setting, TIMP-1−/− BM cells displayed a defective engraftment, showing that TIMP-1 deletion functionally hinders HSC capability of sustaining regular hematopoiesis. Haviernik et al36  have previously performed BM competitive transplantation assays with the use of TIMP-1−/− WBM cells, concluding that TIMP-1 deficiency does not affect HSC engraftment. In our study, we injected 200 000 WBM cells per mouse, whereas Haviernik et al36  transplanted 5 times as many cells. Furthermore, we found that the engraftment rate of TIMP-1−/− BM cells was significantly lower than the WT counterpart at 1 month after transplantation, but it increased over the following 4 weeks. Similarly to Haviernik et al,36  we found that, at 2 months after transplantation, the engraftment of TIMP-1−/− cells did not significantly diverge from the control group. In contrast, we found that, at 12 weeks, the rate of PB chimerism of TIMP-1−/− cells has dropped significantly (P = .014), suggesting that the specific time point at which the engraftment is measured is critical for interpreting TIMP-1 role in hematopoiesis.

In parallel, we found that progenitor-driven, short-term hematopoiesis is actually enhanced in TIMP-1−/− mice, with an increased output of both CFU-Cs and CFU-Ss. This apparent paradox is probably the result of a compensatory mechanism, activated to limit the detrimental effects of an impaired hematopoietic system. Indeed, we found the percentage of KSL progenitors in the spleen to be significantly higher than the control, suggesting that the extramedullary splenic hematopoiesis contributes to rescue the PB phenotype.

Because TIMP-1 is a soluble protein, it still remained unclear whether the impaired function of TIMP-1−/− HSCs was intrinsic to the stem cell compartment or could rather be attributed to the lack of TIMP-1 in the BM niche. To answer this question, we transplanted WT hematopoietic cells into WT and TIMP-1−/− recipients and found no significant differences in the engraftment rates. This suggests that, despite that TIMP-1 is a secreted protein, its influence on HSCs is cell autonomous and intrinsically linked to the genotype of the transplanted cells rather than to the surrounding microenvironment. Similarly, Soloway et al37  demonstrated that TIMP-1−/− mice display the same behavior of the WT counterpart when tested as hosts in metastasis assays, suggesting that TIMP-1 influence on tumor depends exclusively on the TIMP-1 genotype of neoplastic cells rather than the one of the host. Nonetheless, alternative scenarios are still possible. The chronic, rather than acute, lack of TIMP-1 in the BM niche could play a role in the regulation of HSCs, and the general absence of TIMP-1 may be responsible for the hematopoietic phenotype, regardless of the tissue or cell type producing TIMP-1. In addition, the constitutive lack of TIMP-1 could be critical, especially during the early phases of hematopoietic development, rather than during adult life. Thus, we cannot completely rule out an environmental role for TIMP that would be consistent with some of our data.

MMP-2 and MMP-9 have been extensively investigated as key players in HSC trafficking. The release of MMPs from neutrophil granules, secondary to G-CSF mobilization regimens, leads to CXCL12 cleavage, thus favoring the mobilization of HSCs from the BM to the PB.38  Because of the capability of binding pro-MMP-9 and inhibiting its proteolytic activity, we expected TIMP-1 deficiency to enhance HSC mobilization. Nevertheless, the percentage of KSL cells circulating in the PB, in steady-state conditions, did not appear to be affected in TIMP-1−/− mice. Similarly, Haviernik et al36  demonstrated that TIMP-1 deficiency does not influence HSC mobilization in mice undergoing G-CSF treatment, suggesting that other proteases might play a compensatory role.

Our findings indicate that the functional impairment of the stem cell pool in TIMP-1−/− mice is linked to subversion of cell-cycle dynamics. HSCs in TIMP-1 KO mice present an abnormal cell cycle distribution, with a significant decrease in the percentage of quiescent cells in G0. However, despite that an increased percentage of HSCs leaves quiescence and enters G1, a lower percentage of HSCs is in S/G2M, suggesting a defect in the G1/S transition. Failure to efficiently achieve cell-cycle completion may help explain the decline in BM cellularity in KO mice and the impaired engraftment of TIMP-1−/− BM cells, when tested in a competitive setting. These findings suggest that TIMP-1 may play a role in the preservation of HSC quiescence, which is essential to ensure lifelong maintenance of hematopoiesis and to prevent premature exhaustion under conditions of hematopoietic stress, such as transplantations, infections, or chemotherapeutic insults. On the contrary, despite the antiapoptotic role in cancer cell lines that TIMP-1 has been shown to play, HSCs in TIMP-1–deficient mice do not seem to present any evident survival defect. Although we cannot exclude the possibility that a survival defect might arise after a more intense stress, it seems probable that inhibition of apoptosis in cellular pools critical for survival, such as stem cells, would be protected by redundant molecular pathways, capable of compensating for TIMP-1 absence.

Despite intense focus on the signaling pathways activated by TIMP-1, many of the controversies concerning TIMP-1 molecular mechanisms are yet to be reconciled. Among several hypotheses, one of the most intriguing is based on the existence of a specific membrane receptor, the CD63 protein,39  whose binding to TIMP-1 is assumed to lead to proliferation and survival. CD63 was the first-identified tetraspanin, a member of a large family of receptors involved in cell motility, adhesion, and survival.39  Tetraspanins have been shown to form complex networks with other membrane proteins, generating tetraspanin-enriched microdomains. Interestingly, Jung et al17  demonstrated that TIMP-1 contributes to the formation of the ternary complex TIMP-1/CD63/β1-integrin, which inhibits apoptosis in MCF10A breast epithelial cells. Of note, β1-integrin has been previously shown to play a pivotal role in HSCs, by controlling homing and survival.40,41  For instance, Yoshihara et al42  demonstrated the role of THPO/MPL signaling in preserving HSC quiescence through the activation of β1-integrin. Therefore, it seems conceivable that the capability of TIMP-1 in stabilizing the TIMP-1/CD63/β1-integrin complex and keeping β1-integrin in an activated conformation could play a role in preserving HSC quiescence.43  Interestingly, β1-integrin blocking can cause an increase in p53 nuclear levels, thus interfering with proliferation,44  and drug-resistance in B-cell chronic lymphocytic leukemia cells has been suggested to involve p53 inhibition on activation of α4β1-integrin.43  Consistent with these findings, we found that disrupting TIMP-1/CD63 interaction by knocking down TIMP-1 expression compromises HSC quiescence and destabilizes cell cycle dynamics.

The glycoprotein CD44 is an adhesion molecule widely expressed in many cell types, including HSCs. Although CD44 was first described as a key regulator of cell adhesion, it is now widely accepted that its engagement activates signaling cascades involved in cell proliferation, survival, and migration.45,46  Godar et al35  effectively showed that CD44 is fundamental for the proliferation of tumorigenic mammary epithelial cells and demonstrated that its expression is directly inhibited by the tumor suppressor p53, a master regulator of cell proliferation. The investigators showed that, by binding to a noncanonical p53-binding sequence in the CD44 promoter, p53 inhibits CD44 expression and down-regulates the growth-promoting cascade downstream of CD44.35  Interestingly, we found that TIMP-1−/− SPKLS cells not only present cell- cycle dysregulation at the level of the G1/S transition, but they also express significantly lower levels of CD44. We also showed that p53 levels are increased in TIMP-1−/− HSCs. These findings suggest the involvement of the p53/CD44 axis in cell-cycle dysregulation.

On the basis of the aforementioned findings, we propose a hypothetical mechanistic model for TIMP-1 control of proliferation in HSCs. As shown in supplemental Figure 7, the ternary complex TIMP-1/CD63/β1-integrin contributes to maintain β1-integrin activated. When stimulated, β1-integrin keeps p53 levels low, thus promoting cell proliferation through CD44. In contrast, in a TIMP-1−/− model, TIMP-1/CD63/β1-integrin assembling is compromised, resulting in the down-regulation of β1-integrin cascade, with subsequent increase of p53 levels. By binding to the CD44 promoter, p53 represses CD44 expression and causes cell-cycle alteration. Incidentally, because of the role that CD44 plays in CXCL12-driven HSC migration, the decreased expression of CD44 could explain, concomitantly with cell-cycle dysregulation, the decreased responsiveness of TIMP-1−/− HSCs to CXCL12 chemotactic gradients.

TIMP-1 has been proposed as an alternative prognostic marker for the stratification of patients with metastatic breast cancer. Contrary to what the role of TIMP-1 in inhibiting the pro-metastatic factor MMP-9 might suggest, high TIMP-1 levels actually correlate with a worse prognosis and a faster progression of the disease.47,48  This phenomenon seems to be related to the capability of TIMP-1 of promoting cell survival and proliferation of cancer cells, making it an appealing target for novel therapeutic strategies for the treatment of breast cancer, as well as fibrosis secondary to hypertension49  or liver diseases.50,51  Although several compounds are currently under investigation as TIMP-1 inhibitors, the potential side effects of such therapeutic approaches are still controversial and poorly investigated. On the basis of our observations, drugs targeting TIMP-1 could potentially induce complications in terms of hematologic toxicity, advising a cautious use of these compounds. In conclusion, our study shows that TIMP-1 is involved in regulating the LT-HSC cell cycle, thus providing a novel tool for understanding HSC behavior.

The online version of this article contains a data supplement.

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 USC section 1734.

We thank Chris Threeton and Xuejun (Tony) Zhang for help with flow cytometric sorting. We also thank all the members of the Goodell Lab (Baylor College of Medicine), as well as Roberto M. Lemoli, Michele Baccarani, and Sante Tura (University of Bologna) for helpful discussion and support.

This work was supported by a grant from the Italian Association for Leukemias and Lymphomas (BolognAIL; L.R.) and by the National Institutes of Health (grants DK58192, EB005173, and HL081007) and the American Heart Association (grant 0740020N).

National Institutes of Health

Contribution: L.R. designed the research, performed experiments, analyzed the data, and wrote the paper; A.V.E. performed the real-time evaluation of TIMP-1 expression; and M.A.G. designed the research and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Margaret A. Goodell, One Baylor Plaza, N1030, Houston, TX 77030; e-mail: goodell@bcm.edu; or Lara Rossi, Via Massarenti, 9, 40138 Bologna, Italy; e-mail: lararossi79@yahoo.it.

1
Huang
 
X
Cho
 
S
Spangrude
 
GJ
Hematopoietic stem cells: generation and self-renewal.
Cell Death Differ
2007
, vol. 
14
 
11
(pg. 
1851
-
1859
)
2
Arai
 
F
Hirao
 
A
Suda
 
T
Regulation of hematopoiesis and its interaction with stem cell niches.
Int J Hematol
2005
, vol. 
82
 
5
(pg. 
371
-
376
)
3
Yin
 
T
Li
 
L
The stem cell niches in bone.
J Clin Invest
2006
, vol. 
116
 
5
(pg. 
1195
-
1201
)
4
Heissig
 
B
Hattori
 
K
Dias
 
S
, et al. 
Recruitment of stem and progenitor cells from the bone marrow niche requires MMP-9 mediated release of kit-ligand.
Cell
2002
, vol. 
109
 
5
(pg. 
625
-
637
)
5
Raaijmakers
 
MH
Scadden
 
DT
Evolving concepts on the microenvironmental niche for hematopoietic stem cells.
Curr Opin Hematol
2008
, vol. 
15
 
4
(pg. 
301
-
306
)
6
Page-McCaw
 
A
Ewald
 
AJ
Werb
 
Z
Matrix metalloproteinases and the regulation of tissue remodelling.
Nat Rev Mol Cell Biol
2007
, vol. 
8
 
3
(pg. 
221
-
233
)
7
Brew
 
K
Dinakarpandian
 
D
Nagase
 
H
Tissue inhibitors of metalloproteinases: evolution, structure and function.
Biochim Biophys Acta
2000
, vol. 
1477
 
1–2
(pg. 
267
-
283
)
8
Marquez-Curtis
 
LA
Dobrowsky
 
A
Montano
 
J
, et al. 
Matrix metalloproteinase and tissue inhibitors of metalloproteinase secretion by haematopoietic and stromal precursors and their production in normal and leukaemic long-term marrow cultures.
Br J Haematol
2001
, vol. 
115
 
3
(pg. 
595
-
604
)
9
Guedez
 
L
Lim
 
MS
Stetler-Stevenson
 
WG
The role of metalloproteinases and their inhibitors in hematological disorders.
Crit Rev Oncog
1996
, vol. 
7
 
3–4
(pg. 
205
-
225
)
10
Lambert
 
E
Dasse
 
E
Haye
 
B
Petitfrere
 
E
TIMPs as multifacial proteins.
Crit Rev Oncol Hematol
2004
, vol. 
49
 
3
(pg. 
187
-
198
)
11
Chirco
 
R
Liu
 
XW
Jung
 
KK
Kim
 
HR
Novel functions of TIMPs in cell signaling.
Cancer Metastasis Rev
2006
, vol. 
25
 
1
(pg. 
99
-
113
)
12
Wang
 
T
Lv
 
JH
Zhang
 
XF
Li
 
CJ
Han
 
X
Sun
 
YJ
Tissue inhibitor of metalloproteinase-1 protects MCF-7 breast cancer cells from paclitaxel-induced apoptosis by decreasing the stability of cyclin B1.
Int J Cancer
2010
, vol. 
126
 
2
(pg. 
362
-
70
)
13
Stetler-Stevenson
 
WG
Tissue inhibitors of metalloproteinases in cell signaling: metalloproteinase-independent biological activities.
Sci Signal
2008
, vol. 
1
 
27
pg. 
re6
 
14
Nakajima
 
H
Ito
 
M
Smookler
 
DS
, et al. 
TIMP-3 recruits quiescent hematopoietic stem cells into active cell cycle and expands multipotent progenitor pool.
Blood
2010
, vol. 
116
 
22
(pg. 
4474
-
4482
)
15
Liu
 
XW
Taube
 
ME
Jung
 
KK
, et al. 
Tissue inhibitor of metalloproteinase-1 protects human breast epithelial cells from extrinsic cell death: a potential oncogenic activity of tissue inhibitor of metalloproteinase-1.
Cancer Res
2005
, vol. 
65
 
3
(pg. 
898
-
906
)
16
Lambert
 
E
Boudot
 
C
Kadri
 
Z
, et al. 
Tissue inhibitor of metalloproteinases-1 signalling pathway leading to erythroid cell survival.
Biochem J
2003
, vol. 
372
 
pt 3
(pg. 
767
-
774
)
17
Jung
 
KK
Liu
 
XW
Chirco
 
R
Fridman
 
R
Kim
 
HR
Identification of CD63 as a tissue inhibitor of metalloproteinase-1 interacting cell surface protein.
EMBO J
2006
, vol. 
25
 
17
(pg. 
3934
-
3942
)
18
Lambert
 
E
Bridoux
 
L
Devy
 
J
, et al. 
TIMP-1 binding to proMMP-9/CD44 complex localized at the cell surface promotes erythroid cell survival.
Int J Biochem Cell Biol
2009
, vol. 
41
 
5
(pg. 
1102
-
1115
)
19
Chambers
 
SM
Boles
 
NC
Lin
 
KY
, et al. 
Hematopoietic fingerprints: an expression database of stem cells and their progeny.
Cell Stem Cell
2007
, vol. 
1
 
5
(pg. 
578
-
591
)
20
Harrison
 
DE
Competitive repopulation: a new assay for long-term stem cell functional capacity.
Blood
1980
, vol. 
55
 
1
(pg. 
77
-
81
)
21
Challen
 
GA
Boles
 
N
Lin
 
KK
Goodell
 
MA
Mouse hematopoietic stem cell identification and analysis.
Cytometry A
2009
, vol. 
75
 
1
(pg. 
14
-
24
)
22
Goodell
 
MA
Brose
 
K
Paradis
 
G
Conner
 
AS
Mulligan
 
RC
Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo.
J Exp Med
1996
, vol. 
183
 
4
(pg. 
1797
-
1806
)
23
Huttmann
 
A
Liu
 
SL
Boyd
 
AW
Li
 
CL
Functional heterogeneity within rhodamine123(lo) Hoechst33342(lo/sp) primitive hemopoietic stem cells revealed by pyronin Y.
Exp Hematol
2001
, vol. 
29
 
9
(pg. 
1109
-
1116
)
24
Eliasson
 
P
Rehn
 
M
Hammar
 
P
, et al. 
Hypoxia mediates low cell-cycle activity and increases the proportion of long-term-reconstituting hematopoietic stem cells during in vitro culture.
Exp Hematol
2010
, vol. 
38
 
4
(pg. 
301
-
310
)pg. 
e302
 
25
Lei
 
W
Liu
 
F
Ness
 
SA
Positive and negative regulation of c-Myb by cyclin D1, cyclin-dependent kinases, and p27 Kip1.
Blood
2005
, vol. 
105
 
10
(pg. 
3855
-
3861
)
26
Qian
 
H
Buza-Vidas
 
N
Hyland
 
CD
, et al. 
Critical role of thrombopoietin in maintaining adult quiescent hematopoietic stem cells.
Cell Stem Cell
2007
, vol. 
1
 
6
(pg. 
671
-
684
)
27
Yamazaki
 
S
Iwama
 
A
Morita
 
Y
Eto
 
K
Ema
 
H
Nakauchi
 
H
Cytokine signaling, lipid raft clustering, and HSC hibernation.
Ann N Y Acad Sci
2007
, vol. 
1106
 (pg. 
54
-
63
)
28
Lapidot
 
T
Dar
 
A
Kollet
 
O
How do stem cells find their way home?
Blood
2005
, vol. 
106
 
6
(pg. 
1901
-
1910
)
29
Avigdor
 
A
Goichberg
 
P
Shivtiel
 
S
, et al. 
CD44 and hyaluronic acid cooperate with SDF-1 in the trafficking of human CD34+ stem/progenitor cells to bone marrow.
Blood
2004
, vol. 
103
 
8
(pg. 
2981
-
2989
)
30
Vermeulen
 
M
Le Pesteur
 
F
Gagnerault
 
MC
Mary
 
JY
Sainteny
 
F
Lepault
 
F
Role of adhesion molecules in the homing and mobilization of murine hematopoietic stem and progenitor cells.
Blood
1998
, vol. 
92
 
3
(pg. 
894
-
900
)
31
Khaldoyanidi
 
S
Denzel
 
A
Zoller
 
M
Requirement for CD44 in proliferation and homing of hematopoietic precursor cells.
J Leukoc Biol
1996
, vol. 
60
 
5
(pg. 
579
-
592
)
32
Ghaffari
 
S
Smadja-Joffe
 
F
Oostendorp
 
R
, et al. 
CD44 isoforms in normal and leukemic hematopoiesis.
Exp Hematol
1999
, vol. 
27
 
6
(pg. 
978
-
993
)
33
Krause
 
DS
Spitzer
 
TR
Stowell
 
CP
The concentration of CD44 is increased in hematopoietic stem cell grafts of patients with acute myeloid leukemia, plasma cell myeloma, and non-Hodgkin lymphoma.
Arch Pathol Lab Med
2010
, vol. 
134
 
7
(pg. 
1033
-
1038
)
34
Krause
 
DS
Lazarides
 
K
von Andrian
 
UH
Van Etten
 
RA
Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells.
Nat Med
2006
, vol. 
12
 
10
(pg. 
1175
-
1180
)
35
Godar
 
S
Ince
 
TA
Bell
 
GW
, et al. 
Growth-inhibitory and tumor-suppressive functions of p53 depend on its repression of CD44 expression.
Cell
2008
, vol. 
134
 
1
(pg. 
62
-
73
)
36
Haviernik
 
P
Diaz
 
MT
Haviernikova
 
E
Tse
 
W
Stetler-Stevenson
 
WG
Bunting
 
KD
Hematopoiesis in mice is extremely resilient to wide variation in TIMP/MMP balance.
Blood Cells Mol Dis
2008
, vol. 
41
 
2
(pg. 
179
-
187
)
37
Soloway
 
PD
Alexander
 
CM
Werb
 
Z
Jaenisch
 
R
Targeted mutagenesis of Timp-1 reveals that lung tumor invasion is influenced by Timp-1 genotype of the tumor but not by that of the host.
Oncogene
1996
, vol. 
13
 
11
(pg. 
2307
-
2314
)
38
Levesque
 
JP
Liu
 
F
Simmons
 
PJ
, et al. 
Characterization of hematopoietic progenitor mobilization in protease-deficient mice.
Blood
2004
, vol. 
104
 
1
(pg. 
65
-
72
)
39
Pols
 
MS
Klumperman
 
J
Trafficking and function of the tetraspanin CD63.
Exp Cell Res
2009
, vol. 
315
 
9
(pg. 
1584
-
1592
)
40
Potocnik
 
AJ
Brakebusch
 
C
Fassler
 
R
Fetal and adult hematopoietic stem cells require beta1 integrin function for colonizing fetal liver, spleen, and bone marrow.
Immunity
2000
, vol. 
12
 
6
(pg. 
653
-
663
)
41
Szilvassy
 
SJ
Meyerrose
 
TE
Ragland
 
PL
Grimes
 
B
Homing and engraftment defects in ex vivo expanded murine hematopoietic cells are associated with downregulation of beta1 integrin.
Exp Hematol
2001
, vol. 
29
 
12
(pg. 
1494
-
1502
)
42
Yoshihara
 
H
Arai
 
F
Hosokawa
 
K
, et al. 
Thrombopoietin/MPL signaling regulates hematopoietic stem cell quiescence and interaction with the osteoblastic niche.
Cell Stem Cell
2007
, vol. 
1
 
6
(pg. 
685
-
697
)
43
de la Fuente
 
MT
Casanova
 
B
Cantero
 
E
, et al. 
Involvement of p53 in alpha4beta1 integrin-mediated resistance of B-CLL cells to fludarabine.
Biochem Biophys Res Commun
2003
, vol. 
311
 
3
(pg. 
708
-
712
)
44
Blundell
 
R
Harrison
 
D
Changes in the expression of p21, p27, and p53 following beta1-integrin blocking in murine Clare cells cultures.
Int J Mol Med Adv Sci
2006
, vol. 
2
 (pg. 
245
-
250
)
45
Bourguignon
 
LY
Hyaluronan-mediated CD44 activation of RhoGTPase signaling and cytoskeleton function promotes tumor progression.
Semin Cancer Biol
2008
, vol. 
18
 
4
(pg. 
251
-
259
)
46
Wallach-Dayan
 
SB
Rubinstein
 
AM
Hand
 
C
Breuer
 
R
Naor
 
D
DNA vaccination with CD44 variant isoform reduces mammary tumor local growth and lung metastasis.
Mol Cancer Ther
2008
, vol. 
7
 
6
(pg. 
1615
-
1623
)
47
Wurtz
 
SO
Schrohl
 
AS
Sorensen
 
NM
, et al. 
Tissue inhibitor of metalloproteinases-1 in breast cancer.
Endocr Relat Cancer
2005
, vol. 
12
 
2
(pg. 
215
-
227
)
48
Wurtz
 
SO
Schrohl
 
AS
Mouridsen
 
H
Brunner
 
N
TIMP-1 as a tumor marker in breast cancer–an update.
Acta Oncol
2008
, vol. 
47
 
4
(pg. 
580
-
590
)
49
Hultstrom
 
M
Leh
 
S
Skogstrand
 
T
Iversen
 
BM
Upregulation of tissue inhibitor of metalloproteases-1 (TIMP-1) and procollagen-N-peptidase in hypertension-induced renal damage.
Nephrol Dial Transplant
2008
, vol. 
23
 
3
(pg. 
896
-
903
)
50
Nie
 
QH
Zhang
 
YF
Xie
 
YM
, et al. 
Correlation between TIMP-1 expression and liver fibrosis in two rat liver fibrosis models.
World J Gastroenterol
2006
, vol. 
12
 
19
(pg. 
3044
-
3049
)
51
Nie
 
QH
Cheng
 
YQ
Xie
 
YM
Zhou
 
YX
Cao
 
YZ
Inhibiting effect of antisense oligonucleotides phosphorthioate on gene expression of TIMP-1 in rat liver fibrosis.
World J Gastroenterol
2001
, vol. 
7
 
3
(pg. 
363
-
369
)
Sign in via your Institution