8 research outputs found

    Lameness Associated with a Possible Neural Problem in the Lower Spinal Cord of Chickens, Gallus Gallus

    Get PDF
    Since the second half of the twentieth century lameness in broiler chickens has been known to be caused by different types of disorders, however the etiology of several type of lameness remain unknown. Because of the intense selection of broilers for rapid growth, some birds are more prone to physiological insults resulting in lameness. This study focuses on possible neural problems in the lower spinal cord associated with lameness in broilers. Broilers were raised in pens with wire floors and provided with food and water ad libitum. Three groups of birds 1) birds displaying a normal gait (Controls), 2) lame birds with normal leg bones (Neural Associated Lameness), and 3) lame birds with femoral head separation/necrosis (Bone Associated Lameness) were selected based upon behavioral observations of gait. Motor neurons in the lateral motor column of the lumbosacral region of birds in the neural associated lame group were observed to have changes in the perikarya of motor neurons. The lumbosacral segment 4 (LS 4) of the spinal cord was selected for analysis, based upon its central location in the lumbosacral region and the highest number of motor neurons when compared to other segments. In the neural associated lame group the motor neurons were more globular in appearance with a 25% to 40% reduction in protrusions from the perikarya when compared to controls. The corticosterone level, an indicator of stress, was increased 3-4 fold in the lame birds when compared to the controls. Additionally, an association between decreased percentage body weight gain and increased corticosterone level in lame birds was observed. Results of this study indicate an association between the reductions of neurites in the motor neurons of the lumbosacral spinal cord and a leg weakness in broilers with normal leg bones

    Central Role of Vasotocin in the Neuroendocrine Regulation of Stress Responses and Food Intake in Chickens, Gallus gallus

    Get PDF
    It is well known that arginine vasotocin (AVT) in birds is involved in physiological homeostasis such as cardiovascular, osmotic regulation as well as reproductive functions. Pertinent to these physiological functions, AVT immunoreactive (-ir) neurons in the hypothalamus have been found associated with hemorrhage, dehydration, oviposition and other physiological regulation. Evidence, however, suggests that AVT also plays significant roles in modulating behavior, memory, stress, and food intake. This dissertation research addresses the latter two neuroendocrine functions of AVT in detail within the chicken brain. First, the functional role of AVT-ir neurons in conjunction with corticotropin releasing hormone (CRH)-ir neurons in the neuroendocrine regulation of stress is elucidated in meat type chickens. The results from research showed that a distinct type of AVT neurons (parvocellular - small sized) in the paraventricular nucleus (PVN) of the hypothalamus is activated in response to acute and chronic stress. Furthermore, in order to facilitate the neuroendocrine regulation of stress in chickens, expression of AVT in the PVN occurs in the latter phase of the stress period compared to earlier activity of CRH when birds are exposed to a stressor. Second, the neuromodulatory role of AVT to activate CRH neurons via vasotocin receptors (V1aR) within the avian brain is addressed. The results suggests that AVT could augment stress response by modulating CRH neuronal activity in a septal nucleus called the nucleus of the hippocampal commissure (NHpC). Third, the functional role of AVT via the V1aR in food intake regulation is characterized. Results show the association of AVT neurons (magnocellular – large sized), in the preoptic and hypothalamic brain structures, and CRH neurons, in the NHpC, following food intake in chickens. Hence, neuroendocrine regulations of AVT neurons in distinct brain structures in the stress response and food intake in chickens are characterized by this dissertation research

    AMP-Activated Protein Kinase Mediates the Effect of Leptin on Avian Autophagy in a Tissue-Specific Manner

    No full text
    Autophagy, a highly conserved intracellular self-digestion process, plays an integral role in maintaining cellular homeostasis. Although emerging evidence indicate that the endocrine system regulates autophagy in mammals, there is still a scarcity of information on autophagy in avian (non-mammalian) species. Here, we show that intracerebroventricular administration of leptin reduces feed intake, modulates the expression of feeding-related hypothalamic neuropeptides, activates leptin receptor and signal transducer and activator of transcription (Ob-Rb/STAT) pathway, and significantly increases the expression of autophagy-related proteins (Atg3, Atg5, Atg7, beclin1, and LC3B) in chicken hypothalamus, liver, and muscle. Similarly, leptin treatment activates Ob-Rb/STAT pathway and increased the expression of autophagy-related markers in chicken hypothalamic organotypic cultures, muscle (QM7) and hepatocyte (Sim-CEL) cell cultures as well as in Chinese Hamster Ovary (CHO-K1) cells-overexpressing chicken Ob-Rb and STAT3. To define the downstream mediator(s) of leptin's effects on autophagy, we determined the role of the master energy sensor AMP-activated protein kinase (AMPK). Leptin treatment significantly increased the phosphorylated levels of AMPKα1/2 at Thr172 site in chicken hypothalamus and liver, but not in muscle. Likewise, AMPKα1/2 was activated by leptin in chicken hypothalamic organotypic culture and Sim-CEL, but not in QM7 cells. Blocking AMPK activity by compound C reverses the autophagy-inducing effect of leptin. Together, these findings indicate that AMPK mediates the effect of leptin on chicken autophagy in a tissue-specific manner

    Data_Sheet_1_AMP-Activated Protein Kinase Mediates the Effect of Leptin on Avian Autophagy in a Tissue-Specific Manner.DOCX

    No full text
    <p>Autophagy, a highly conserved intracellular self-digestion process, plays an integral role in maintaining cellular homeostasis. Although emerging evidence indicate that the endocrine system regulates autophagy in mammals, there is still a scarcity of information on autophagy in avian (non-mammalian) species. Here, we show that intracerebroventricular administration of leptin reduces feed intake, modulates the expression of feeding-related hypothalamic neuropeptides, activates leptin receptor and signal transducer and activator of transcription (Ob-Rb/STAT) pathway, and significantly increases the expression of autophagy-related proteins (Atg3, Atg5, Atg7, beclin1, and LC3B) in chicken hypothalamus, liver, and muscle. Similarly, leptin treatment activates Ob-Rb/STAT pathway and increased the expression of autophagy-related markers in chicken hypothalamic organotypic cultures, muscle (QM7) and hepatocyte (Sim-CEL) cell cultures as well as in Chinese Hamster Ovary (CHO-K1) cells-overexpressing chicken Ob-Rb and STAT3. To define the downstream mediator(s) of leptin's effects on autophagy, we determined the role of the master energy sensor AMP-activated protein kinase (AMPK). Leptin treatment significantly increased the phosphorylated levels of AMPKα1/2 at Thr172 site in chicken hypothalamus and liver, but not in muscle. Likewise, AMPKα1/2 was activated by leptin in chicken hypothalamic organotypic culture and Sim-CEL, but not in QM7 cells. Blocking AMPK activity by compound C reverses the autophagy-inducing effect of leptin. Together, these findings indicate that AMPK mediates the effect of leptin on chicken autophagy in a tissue-specific manner.</p

    Data_Sheet_2_AMP-Activated Protein Kinase Mediates the Effect of Leptin on Avian Autophagy in a Tissue-Specific Manner.DOCX

    No full text
    <p>Autophagy, a highly conserved intracellular self-digestion process, plays an integral role in maintaining cellular homeostasis. Although emerging evidence indicate that the endocrine system regulates autophagy in mammals, there is still a scarcity of information on autophagy in avian (non-mammalian) species. Here, we show that intracerebroventricular administration of leptin reduces feed intake, modulates the expression of feeding-related hypothalamic neuropeptides, activates leptin receptor and signal transducer and activator of transcription (Ob-Rb/STAT) pathway, and significantly increases the expression of autophagy-related proteins (Atg3, Atg5, Atg7, beclin1, and LC3B) in chicken hypothalamus, liver, and muscle. Similarly, leptin treatment activates Ob-Rb/STAT pathway and increased the expression of autophagy-related markers in chicken hypothalamic organotypic cultures, muscle (QM7) and hepatocyte (Sim-CEL) cell cultures as well as in Chinese Hamster Ovary (CHO-K1) cells-overexpressing chicken Ob-Rb and STAT3. To define the downstream mediator(s) of leptin's effects on autophagy, we determined the role of the master energy sensor AMP-activated protein kinase (AMPK). Leptin treatment significantly increased the phosphorylated levels of AMPKα1/2 at Thr172 site in chicken hypothalamus and liver, but not in muscle. Likewise, AMPKα1/2 was activated by leptin in chicken hypothalamic organotypic culture and Sim-CEL, but not in QM7 cells. Blocking AMPK activity by compound C reverses the autophagy-inducing effect of leptin. Together, these findings indicate that AMPK mediates the effect of leptin on chicken autophagy in a tissue-specific manner.</p

    Data_Sheet_4_AMP-Activated Protein Kinase Mediates the Effect of Leptin on Avian Autophagy in a Tissue-Specific Manner.DOCX

    No full text
    <p>Autophagy, a highly conserved intracellular self-digestion process, plays an integral role in maintaining cellular homeostasis. Although emerging evidence indicate that the endocrine system regulates autophagy in mammals, there is still a scarcity of information on autophagy in avian (non-mammalian) species. Here, we show that intracerebroventricular administration of leptin reduces feed intake, modulates the expression of feeding-related hypothalamic neuropeptides, activates leptin receptor and signal transducer and activator of transcription (Ob-Rb/STAT) pathway, and significantly increases the expression of autophagy-related proteins (Atg3, Atg5, Atg7, beclin1, and LC3B) in chicken hypothalamus, liver, and muscle. Similarly, leptin treatment activates Ob-Rb/STAT pathway and increased the expression of autophagy-related markers in chicken hypothalamic organotypic cultures, muscle (QM7) and hepatocyte (Sim-CEL) cell cultures as well as in Chinese Hamster Ovary (CHO-K1) cells-overexpressing chicken Ob-Rb and STAT3. To define the downstream mediator(s) of leptin's effects on autophagy, we determined the role of the master energy sensor AMP-activated protein kinase (AMPK). Leptin treatment significantly increased the phosphorylated levels of AMPKα1/2 at Thr172 site in chicken hypothalamus and liver, but not in muscle. Likewise, AMPKα1/2 was activated by leptin in chicken hypothalamic organotypic culture and Sim-CEL, but not in QM7 cells. Blocking AMPK activity by compound C reverses the autophagy-inducing effect of leptin. Together, these findings indicate that AMPK mediates the effect of leptin on chicken autophagy in a tissue-specific manner.</p

    Data_Sheet_3_AMP-Activated Protein Kinase Mediates the Effect of Leptin on Avian Autophagy in a Tissue-Specific Manner.DOCX

    No full text
    <p>Autophagy, a highly conserved intracellular self-digestion process, plays an integral role in maintaining cellular homeostasis. Although emerging evidence indicate that the endocrine system regulates autophagy in mammals, there is still a scarcity of information on autophagy in avian (non-mammalian) species. Here, we show that intracerebroventricular administration of leptin reduces feed intake, modulates the expression of feeding-related hypothalamic neuropeptides, activates leptin receptor and signal transducer and activator of transcription (Ob-Rb/STAT) pathway, and significantly increases the expression of autophagy-related proteins (Atg3, Atg5, Atg7, beclin1, and LC3B) in chicken hypothalamus, liver, and muscle. Similarly, leptin treatment activates Ob-Rb/STAT pathway and increased the expression of autophagy-related markers in chicken hypothalamic organotypic cultures, muscle (QM7) and hepatocyte (Sim-CEL) cell cultures as well as in Chinese Hamster Ovary (CHO-K1) cells-overexpressing chicken Ob-Rb and STAT3. To define the downstream mediator(s) of leptin's effects on autophagy, we determined the role of the master energy sensor AMP-activated protein kinase (AMPK). Leptin treatment significantly increased the phosphorylated levels of AMPKα1/2 at Thr172 site in chicken hypothalamus and liver, but not in muscle. Likewise, AMPKα1/2 was activated by leptin in chicken hypothalamic organotypic culture and Sim-CEL, but not in QM7 cells. Blocking AMPK activity by compound C reverses the autophagy-inducing effect of leptin. Together, these findings indicate that AMPK mediates the effect of leptin on chicken autophagy in a tissue-specific manner.</p
    corecore