Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Susan Mackem is active.

Publication


Featured researches published by Susan Mackem.


Developmental Cell | 2010

Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels.

Christa Maes; Tatsuya Kobayashi; Martin K. Selig; Sophie Torrekens; Sanford I. Roth; Susan Mackem; Geert Carmeliet; Henry M. Kronenberg

During endochondral bone development, the first osteoblasts differentiate in the perichondrium surrounding avascular cartilaginous rudiments; the source of trabecular osteoblasts inside the later bone is, however, unknown. Here, we generated tamoxifen-inducible transgenic mice bred to Rosa26R-LacZ reporter mice to follow the fates of stage-selective subsets of osteoblast lineage cells. Pulse-chase studies showed that osterix-expressing osteoblast precursors, labeled in the perichondrium prior to vascular invasion of the cartilage, give rise to trabecular osteoblasts, osteocytes, and stromal cells inside the developing bone. Throughout the translocation, some precursors were found to intimately associate with invading blood vessels, in pericyte-like fashion. A similar coinvasion occurs during endochondral healing of bone fractures. In contrast, perichondrial mature osteoblasts did not exhibit perivascular localization and remained in the outer cortex of developing bones. These findings reveal the specific involvement of immature osteoblast precursors in the coupled vascular and osteogenic transformation essential to endochondral bone development and repair.


The EMBO Journal | 1998

Soluble dominant-negative receptor uncovers essential roles for fibroblast growth factors in multi-organ induction and patterning

Giulia Celli; William J. LaRochelle; Susan Mackem; Richard Sharp; Glenn Merlino

Despite a wealth of experimental data implicating fibroblast growth factor (FGF) signaling in various developmental processes, genetic inactivation of individual genes encoding specific FGFs or their receptors (FGFRs) has generally failed to demonstrate their role in vertebrate organogenesis due to early embryonic lethality or functional redundancy. Here we show that broad mid‐gestational expression of a novel secreted kinase‐deficient receptor, specific for a defined subset of the FGF superfamily, caused agenesis or severe dysgenesis of kidney, lung, specific cutaneous structures, exocrine and endocrine glands, and craniofacial and limb abnormalities reminiscent of human skeletal disorders associated with FGFR mutations. Analysis of diagnostic molecular markers revealed that this soluble dominant‐negative mutant disrupted early inductive signaling in affected tissues, indicating that FGF signaling is required for growth and patterning in a broad array of organs and in limbs. In contrast, transgenic mice expressing a membrane‐tethered kinase‐deficient FGFR were viable. Our results demonstrate that secreted FGFR mutants are uniquely effective as dominant‐negative agents in vivo, and suggest that related soluble receptor isoforms expressed in wild‐type mouse embryos may help regulate FGF activity during normal development.


Developmental Cell | 2008

Uncoupling Sonic hedgehog control of pattern and expansion of the developing limb bud.

Jianjian Zhu; Eiichiro Nakamura; Minh-Thanh Nguyen; Xiaozhong Bao; Haruhiko Akiyama; Susan Mackem

Sonic hedgehog (Shh), which regulates proliferation in many contexts, functions as a limb morphogen to specify a distinct pattern of digits. How Shhs effects on cell number relate to its role in specifying digit identity is unclear. Deleting the mouse Shh gene at different times using a conditional Cre line, we find that Shh functions to control limb development in two phases: a very transient, early patterning phase regulating digit identity, and an extended growth-promoting phase during which the digit precursor mesenchyme expands and becomes recruited into condensing digit primordia. Our analysis reveals an unexpected alternating anterior-posterior sequence of normal mammalian digit formation. The progressive loss of digits upon successively earlier Shh removal mirrors this alternating sequence and highlights Shhs role in cell expansion to produce the normal digit complement.


Proceedings of the National Academy of Sciences of the United States of America | 2007

Indian Hedgehog produced by postnatal chondrocytes is essential for maintaining a growth plate and trabecular bone

Yukiko Maeda; Eiichiro Nakamura; Minh-Thanh Nguyen; Larry J. Suva; Frances L. Swain; Mohammed S. Razzaque; Susan Mackem; Beate Lanske

Indian hedgehog (Ihh) is essential for chondrocyte and osteoblast proliferation/differentiation during prenatal endochondral bone formation. The early lethality of various Ihh-ablated mutant mice, however, prevented further analysis of its role in postnatal bone growth and development. In this study, we describe the generation and characterization of a mouse model in which the Ihh gene was successfully ablated from postnatal chondrocytes in a temporal/spatial-specific manner; postnatal deletion of Ihh resulted in loss of columnar structure, premature vascular invasion, and formation of ectopic hypertrophic chondrocytes in the growth plate. Furthermore, destruction of the articular surface in long bones and premature fusion of growth plates of various endochondral bones was evident, resulting in dwarfism in mutant mice. More importantly, these mutant mice exhibited continuous loss of trabecular bone over time, which was accompanied by reduced Wnt signaling in the osteoblastic cells. These results demonstrate, for the first time, that postnatal chondrocyte-derived Ihh is essential for maintaining the growth plate and articular surface and is required for sustaining trabecular bone and skeletal growth.


Development | 2008

Indian hedgehog signals independently of PTHrP to promote chondrocyte hypertrophy

Kinglun Kingston Mak; Henry M. Kronenberg; Pao-Tien Chuang; Susan Mackem; Yingzi Yang

Chondrocyte hypertrophy is an essential process required for endochondral bone formation. Proper regulation of chondrocyte hypertrophy is also required in postnatal cartilage homeostasis. Indian hedgehog (Ihh) and PTHrP signaling play crucial roles in regulating the onset of chondrocyte hypertrophy by forming a negative feedback loop, in which Ihh signaling regulates chondrocyte hypertrophy by controlling PTHrP expression. To understand whether there is a PTHrP-independent role of Ihh signaling in regulating chondrocyte hypertrophy, we have both activated and inactivated Ihh signaling in the absence of PTHrP during endochondral skeletal development. We found that upregulating Ihh signaling in the developing cartilage by treating PTHrP-/- limb explants with sonic hedgehog (Shh) protein in vitro, or overexpressing Ihh in the cartilage of PTHrP-/- embryos or inactivating patched 1 (Ptch1), a negative regulator of hedgehog (Hh) signaling, accelerated chondrocyte hypertrophy in the PTHrP-/- embryos. Conversely, when Hh signaling was blocked by cyclopamine or by removing Smoothened (Smo), a positive regulator of Hh signaling, chondrocyte hypertrophy was delayed in the PTHrP-/- embryo. Furthermore, we show that upregulated Hh signaling in the postnatal cartilage led to accelerated chondrocyte hypertrophy during secondary ossification, which in turn caused reduction of joint cartilage. Our results revealed a novel role of Ihh signaling in promoting chondrocyte hypertrophy independently of PTHrP, which is particularly important in postnatal cartilage development and homeostasis. In addition, we found that bone morphogenetic protein (Bmp) and Wnt/β-catenin signaling in the cartilage may both mediate the effect of upregulated Ihh signaling in promoting chondrocyte hypertrophy.


Developmental Dynamics | 2006

Kinetics of tamoxifen‐regulated Cre activity in mice using a cartilage‐specific CreERT to assay temporal activity windows along the proximodistal limb skeleton

Eiichiro Nakamura; Minh-Thanh Nguyen; Susan Mackem

Cartilage differentiation occurs over a broad time range from early embryonic development, when the mesenchymal condensations that give rise to cartilage models for future bone first appear, and continuing through adult life, when there is ongoing maintenance of articular joint surfaces and re‐activation of cartilage formation after fracture. The chondrogenic response also figures in the pathogenesis of degenerative and inflammatory joint diseases. We have generated a transgenic line expressing tamoxifen‐dependent Cre recombinase that gives efficient recombination in the chondrogenic lineage, both during embryogenesis and postnatally, and provides a valuable tool for analysis of gene function selectively in chondrogenic cells using conditional genetic approaches. Because the cartilage model of the limb skeleton forms progressively in a proximodistal order during discrete, well‐defined time periods, evaluation of the spatial extent of tamoxifen‐induced recombination along the limb axis during these time windows has also enabled us to examine the pharmacokinetics of single‐dose tamoxifen injections during pregnancy. Developmental Dynamics 235:2603–2612, 2006.


PLOS Genetics | 2010

Distinct Roles of Hand2 in Initiating Polarity and Posterior Shh Expression during the Onset of Mouse Limb Bud Development

Antonella Galli; Dimitri Robay; Marco Osterwalder; Xiaozhong Bao; Jean-Denis Bénazet; Muhammad Tariq; Renato Paro; Susan Mackem; Rolf Zeller

The polarization of nascent embryonic fields and the endowment of cells with organizer properties are key to initiation of vertebrate organogenesis. One such event is antero-posterior (AP) polarization of early limb buds and activation of morphogenetic Sonic Hedgehog (SHH) signaling in the posterior mesenchyme, which in turn promotes outgrowth and specifies the pentadactylous autopod. Inactivation of the Hand2 transcriptional regulator from the onset of mouse forelimb bud development disrupts establishment of posterior identity and Shh expression, which results in a skeletal phenotype identical to Shh deficient limb buds. In wild-type limb buds, Hand2 is part of the protein complexes containing Hoxd13, another essential regulator of Shh activation in limb buds. Chromatin immunoprecipitation shows that Hand2-containing chromatin complexes are bound to the far upstream cis-regulatory region (ZRS), which is specifically required for Shh expression in the limb bud. Cell-biochemical studies indicate that Hand2 and Hoxd13 can efficiently transactivate gene expression via the ZRS, while the Gli3 repressor isoform interferes with this positive transcriptional regulation. Indeed, analysis of mouse forelimb buds lacking both Hand2 and Gli3 reveals the complete absence of antero-posterior (AP) polarity along the entire proximo-distal axis and extreme digit polydactyly without AP identities. Our study uncovers essential components of the transcriptional machinery and key interactions that set-up limb bud asymmetry upstream of establishing the SHH signaling limb bud organizer.


Journal of Bone and Mineral Research | 2011

IGF-1R signaling in chondrocytes modulates growth plate development by interacting with the PTHrP/Ihh pathway.

Yongmei Wang; Zhiqiang Cheng; Hashem Elalieh; Eiichiro Nakamura; Minh Thanh Nguyen; Susan Mackem; Thomas L. Clemens; Daniel D. Bikle; Wenhan Chang

Systemic derangements and perinatal death of generalized insulin‐like growth factor 1 (IGF‐1) and IGF‐1 receptor (IGF‐1R) knockout mice preclude definitive assessment of IGF‐1R actions in growth‐plate (GP) chondrocytes. We generated cartilage‐specific Igf1r knockout (CartIgf1r−/−) mice to investigate local control of chondrocyte differentiation in the GP by this receptor. These mice died shortly after birth and showed disorganized chondrocyte columns, delayed ossification and vascular invasion, decreased cell proliferation, increased apoptosis, and increased expression of parathyroid hormone–related protein (Pthrp) RNA and protein in their GPs. The increased Pthrp expression in the knockout GPs likely was due to an increase in gene transcription, as determined by the increased activity of a LacZ reporter that was inserted downstream of the endogenous PTHrP promoter and bred into the knockout mice. To circumvent the early death of CartIgf1r−/− mice and investigate the role of IGF‐1R during postnatal growth, we made tamoxifen (Tam)–inducible, cartilage‐specific Igf1r knockout (TamCartIgf1r−/−) mice. At 2 weeks of age and 7 to 8 days after Tam injection, the TamCartIgf1r−/− mice showed growth retardation with a disorganized GP, reduced chondrocyte proliferation, decreased type 2 collagen and Indian Hedgehog (Ihh) expression, but increased expression of PTHrP. Consistent with in vivo observations, in vitro knockout of the Igf1r gene by adenoviral expression of Cre recombinase suppressed cell proliferation, promoted apoptosis, and increased Pthrp expression. Our data indicate that the IGF‐1R in chondrocytes controls cell growth, survival, and differentiation in embryonic and postnatal GPs in part by suppression of Pthrp expression.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Parathyroid hormone/parathyroid hormone-related protein receptor signaling is required for maintenance of the growth plate in postnatal life

Takao Hirai; Andrei S. Chagin; Tatsuya Kobayashi; Susan Mackem; Henry M. Kronenberg

Parathyroid hormone (PTH)-related protein (PTHrP), regulated by Indian hedgehog and acting through the PTH/PTHrP receptor (PPR), is crucial for normal cartilage development. These observations suggest a possible role of PPR signaling in the postnatal growth plate; however, the role of PPR signaling in postnatal chondrocytes is unknown. In this study, we have generated tamoxifen-inducible and cartilage-specific PPR KO mice to evaluate the physiological role of PPR signaling in postnatal chondrocytes. We found that inactivation of the PPR in chondrocytes postnatally leads to accelerated differentiation of chondrocytes, followed by disappearance of the growth plate. We also observed an increase of TUNEL-positive cells and activities of caspase-3 and caspase-9 in the growth plate, along with a decrease in phosphorylation of Bad at Ser155 in postnatal PPR KO mice. Administration of a low-phosphate diet, which prevents apoptosis of chondrocytes, prevented the disappearance of the growth plate. Taken together, these observations suggest that the major consequences of PPR activation are similar in both the fetal and postnatal growth plates. Moreover, chondrocyte apoptosis through the activation of a mitochondrial pathway may be involved in the process of premature disappearance of the growth plate by postnatal inactivation of the PPR in chondrocytes.


Proceedings of the National Academy of Sciences of the United States of America | 2010

A mouse model of chondrocyte-specific somatic mutation reveals a role for Ext1 loss of heterozygosity in multiple hereditary exostoses

Kazu Matsumoto; Fumitoshi Irie; Susan Mackem; Yu Yamaguchi

Multiple hereditary exostoses (MHE) is one of the most common skeletal dysplasias, exhibiting the formation of multiple cartilage-capped bony protrusions (osteochondroma) and characteristic bone deformities. Individuals with MHE carry heterozygous loss-of-function mutations in Ext1 or Ext2, genes which together encode an enzyme essential for heparan sulfate synthesis. Despite the identification of causative genes, the pathogenesis of MHE remains unclear, especially with regard to whether osteochondroma results from loss of heterozygosity of the Ext genes. Hampering elucidation of the pathogenic mechanism of MHE, both Ext1+/− and Ext2+/− heterozygous mutant mice, which mimic the genetic status of human MHE, are highly resistant to osteochondroma formation, especially in long bones. To address these issues, we created a mouse model in which Ext1 is stochastically inactivated in a chondrocyte-specific manner. We show that these mice develop multiple osteochondromas and characteristic bone deformities in a pattern and a frequency that are almost identical to those of human MHE, suggesting a role for Ext1 LOH in MHE. Surprisingly, however, genotyping and fate mapping analyses reveal that chondrocytes constituting osteochondromas are mixtures of mutant and wild-type cells. Moreover, osteochondromas do not possess many typical neoplastic properties. Together, our results suggest that inactivation of Ext1 in a small fraction of chondrocytes is sufficient for the development of osteochondromas and other skeletal defects associated with MHE. Because the observed osteochondromas in our mouse model do not arise from clonal growth of chondrocytes, they cannot be considered true neoplasms.

Collaboration


Dive into the Susan Mackem's collaboration.

Top Co-Authors

Avatar

Jianjian Zhu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Eiichiro Nakamura

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Minh-Thanh Nguyen

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Xiaozhong Bao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mark Lewandoski

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Vladimir Knezevic

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gordon L. Hager

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kathleen A. Mahon

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lino Tessarollo

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge